US9187521B2 - Tubulysins and processes for preparing - Google Patents

Tubulysins and processes for preparing Download PDF

Info

Publication number
US9187521B2
US9187521B2 US12/739,579 US73957908A US9187521B2 US 9187521 B2 US9187521 B2 US 9187521B2 US 73957908 A US73957908 A US 73957908A US 9187521 B2 US9187521 B2 US 9187521B2
Authority
US
United States
Prior art keywords
alkyl
formula
aryl
alkenyl
optionally substituted
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related, expires
Application number
US12/739,579
Other versions
US20100240701A1 (en
Inventor
Iontcho Radoslavov Vlahov
Christopher Paul Leamon
Yu Wang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Endocyte Inc
Original Assignee
Endocyte Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Endocyte Inc filed Critical Endocyte Inc
Priority to US12/739,579 priority Critical patent/US9187521B2/en
Assigned to ENDOCYTE, INC. reassignment ENDOCYTE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: LEAMON, CHRISTOPHER PAUL, VLAHOV, IONTCHO RADOSLAVOV, WANG, YU
Publication of US20100240701A1 publication Critical patent/US20100240701A1/en
Application granted granted Critical
Publication of US9187521B2 publication Critical patent/US9187521B2/en
Expired - Fee Related legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/02Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing at least one abnormal peptide link
    • C07K5/021Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing at least one abnormal peptide link containing the structure -NH-(X)n-C(=0)-, n being 5 or 6; for n > 6, classification in C07K5/06 - C07K5/10, according to the moiety having normal peptide bonds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06139Dipeptides with the first amino acid being heterocyclic

Definitions

  • the invention described herein pertains to tubulysins and tubulysin analogs, and processes for preparing tubulysins and tubulysin analogs.
  • Tubulysins are a group of powerful inhibitors of tubulin polymerization. Tubulysins are useful in treating diseases and disease states that include pathogenic cell populations, such as cancer. Generally, tubulysins are linear tetrapeptides consisting of N-methyl pipecolic acid (Mep), isoleucine (Ile), an unnatural aminoacid called tubuvalin (Tuv), and either an unnatural aminoacid called tubutyrosine (Tut, an analog of tyrosine) or an unnatural aminoacid called tubuphenylalanine (Tup, an analog of phenylalanine), as shown in the following table:
  • one species Archangium gephyra , produces as the main component factors tubulysins A, B, C, G, and I, each of which may be identified by its including the Tut residue.
  • another species Angiococcus disciformis , produces as the main component factors tubulysins D, E, F, and H, each of which may be identified by its including the Tup residue.
  • tubulysins Such bacterial fermentations are convenient sources of tubulysins.
  • mycobacteria produce only certain tubulysins, and/or mixtures of tubulysins, processes are needed for interconverting those tubulysins to the desired factors for medicinal and pharmacological uses.
  • processes are needed for preparing novel tubulysins, tubulysin analogs, and tubulysin derivatives for medicinal and pharmacological uses.
  • processes for preparing tubulysins are also described herein are analogs and derivatives of tubulysins.
  • processes are described for preparing one or more tubulysins from a mixture of tubulysins, such as a mixture of tubulysins produced by fermentation or some other process.
  • processes are described herein for preparing a mixture of tubulysins from one or more tubulysins.
  • processes are described herein for converting one tubulysin into another tubulysin.
  • processes are described herein for converting one or more, or a mixture of tubulysins into one or more, or a mixture of tubulysin analogs.
  • tubulysin refers both collectively and individually to the naturally occurring tubulysin, and the analogs and derivatives of tubulysins described herein, or that may be prepared from the processes described herein.
  • novel tubulysins, tubulysin analogs, and tubulysin derivatives are described herein along with processes for preparing such novel tubulysins, tubulysin analogs, and tubulysin derivatives.
  • the processes include treating one or more tubulysins with an acid to prepare an intermediate.
  • the processes include the step of subsequently reacting the intermediate with a reagent to prepare a mixture of tubulysins from a different mixture of tubulysins, a single tubulysin from a mixture of tubulysins, a mixture of tubulysins from a single tubulysin, or a single tubulysins from a different tubulysin.
  • the intermediate is a compound of formulae (1a), (1b), or (1c):
  • R, R 1 , S, T, U, V, W, Y, and Z are as described hereinbelow in the various embodiments, aspects, and variations thereof.
  • intermediate compounds of formulae (1a), (1b), and (1c) may form salts, such as salts with the residual acid conjugate base.
  • iminium intermediates such as those of formulae (1a), (1b), and (1c) may also be in equilibrium with the corresponding acyl aminal, where the residual acid conjugate base adds to the iminium intermediate.
  • the iminium intermediates may form solvates or hydrates, each of which may be in equilibrium with the iminium intermediates.
  • nucleophiles such as RCN, RXH and the corresponding anions and salts thereof react with iminium intermediates regardless of whether the iminium is in a salt, hydrate, solvate or acylaminal form to prepare the compounds described herein, such as the compounds of formulae (2a), (2b), and (2c), respectively:
  • R, R 1 , R 10 , S, T, U, V, W, Y, and Z are as described hereinbelow in the various embodiments, aspects, and variations thereof.
  • tubulysins described herein refer generally to tetrapeptide compounds of the formula
  • n 1-3;
  • V is H, OR 2 , or halo
  • W is H, OR 2 , or alkyl, where R 2 is independently selected in each instance from H, alkyl, and C(O)R 3 , where R 3 is alkyl, cycloalkyl, alkenyl, aryl, or arylalkyl, each of which is optionally substituted; providing that R 2 is not H when both V and W are OR 2 ; or V and W are taken together with the attached carbon to form a carbonyl;
  • Z is alkyl and Y is O; or Z is alkyl or C(O)R 4 , and Y is absent, where R 4 is alkyl, CF 3 , or aryl;
  • R 1 is H, or R 1 represents 1 to 3 substituents selected from halo, nitro, carboxylate or a derivative thereof, cyano, hydroxyl, alkyl, haloalkyl, alkoxy, haloalkoxy, phenol protecting groups, prodrug moieties, and OR 6 , where R 6 is optionally substituted aryl, C(O)R 7 , P(O)(OR 8 ) 2 , or SO 3 R 8 , where R 7 and R 8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R 8 is a metal cation; and
  • R is OH or a leaving group, or R forms a carboxylic acid derivative.
  • Z is methyl.
  • R 1 is H.
  • R 1 is OR 6 at C(4), where R 6 is H, alkyl, or COR 7 .
  • V is H, and W is OC(O)R 3 .
  • Illustrative leaving groups include, but are not limited to halides, sulfonates, such as triflates, and the like, optionally substituted phenoxy, such as pentafluorophenoxy and the like, intermediates formed from ester forming or amide forming reagents, such as isobutyl chloroformate, DCC, HOBt, EDC, PyBOP, BOP, BOP-Cl, and the like.
  • Illustrative carboxylic acid derivatives include, but are not limited to, esters, amides, imides, acylhydrazides, nitriles, and optionally substituted variations thereof.
  • tubulysins of the following general formula are described
  • n 1-3;
  • V is H, OR 2 , or halo
  • W is H, OR 2 , or alkyl, where R 2 is independently selected in each instance from H, alkyl, or C(O)R 3 , where R 3 is alkyl, alkenyl or aryl, providing that R 2 is not H when both V and W are OR 2 ; or V and W are taken together with the attached carbon to form a carbonyl;
  • Z is alkyl or C(O)R 4 , where R 4 is alkyl, CF 3 , or aryl;
  • T is H or OR 6 , where R 6 is H, alkyl, aryl, COR 7 , P(O)(OR 8 ) 2 , or SO 3 R 8 , where R 7 and R 8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R 8 is a metal cation, or R 6 is a phenol protecting group, or a prodrug moiety;
  • S and U are each independently selected from the group consisting of H, halo, nitro, cyano, alkyl, haloalkyl, alkoxy, and haloalkoxy;
  • R is OH or a leaving group, or R forms a carboxylic acid derivative.
  • natural tubulysins and the corresponding analogs and derivatives thereof are described.
  • Such natural tubulysins are generally linear tetrapeptides consisting of N-methyl pipecolic acid (Mep), isoleucine (Ile), an unnatural aminoacid called tubuvalin (Tuv), and either an unnatural aminoacid called tubutyrosine (Tut, an analog of tyrosine) or an unnatural aminoacid called tubuphenylalanine (Tup, an analog of phenylalanine).
  • naturally occurring tubulysins, and analogs and derivatives thereof, of the following general formula are described
  • R, R 1 , and R 10 are as described in the various embodiments herein.
  • a first tubulysin or alternatively a mixture of tubulysins, is converted into a second tubulysin by preparing an intermediate compound of formula (1a), wherein
  • n 1-3;
  • V is H, OR 2 , or halo
  • W is H, OR 2 , or alkyl, where R 2 is independently selected in each instance from H, alkyl, and C(O)R 3 , where R 3 is alkyl, cycloalkyl, alkenyl, aryl, or arylalkyl, each of which is optionally substituted; providing that R 2 is not H when both V and W are OR 2 ; or V and W are taken together with the attached carbon to form a carbonyl;
  • Z is alkyl and Y is O; or Z is alkyl or C(O)R 4 , and Y is absent, where R 4 is alkyl, CF 3 , or aryl;
  • R 1 is H, or R 1 represents 1 to 3 substituents selected from halo, nitro, carboxylate or a derivative thereof, cyano, hydroxyl, alkyl, haloalkyl, alkoxy, haloalkoxy, phenol protecting groups, prodrug moieties, and OR 6 , where R 6 is optionally substituted aryl, C(O)R 7 , P(O)(OR 8 ) 2 , or SO 3 R 8 , where R 7 and R 8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R 8 is a metal cation; and
  • R is OH or a leaving group, or R forms a carboxylic acid derivative.
  • the intermediate is prepared by mixing a tubulysin or mixture of tubulysins of formula (2b) with an acid under substantially anhydrous conditions:
  • n 1-3;
  • V is H, OR 2 , or halo
  • W is H, OR 2 , or alkyl, where R 2 is independently selected in each instance from H, alkyl, and C(O)R 3 , where R 3 is alkyl, cycloalkyl, alkenyl, aryl, or arylalkyl, each of which is optionally substituted; providing that R 2 is not H when both V and W are OR 2 ; or V and W are taken together with the attached carbon to form a carbonyl;
  • Z is alkyl and Y is O; or Z is alkyl or C(O)R 4 , and Y is absent, where R 4 is alkyl, CF 3 , or aryl;
  • R 1 is H, or R 1 represents 1 to 3 substituents selected from halo, nitro, carboxylate or a derivative thereof, cyano, hydroxyl, alkyl, haloalkyl, alkoxy, haloalkoxy, phenol protecting groups, prodrug moieties, and OR 6 , where R 6 is optionally substituted aryl, C(O)R 7 , P(O)(OR 8 ) 2 , or SO 3 R 8 , where R 7 and R 8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R 8 is a metal cation; and
  • R is OH or a leaving group, or R forms a carboxylic acid derivative.
  • the intermediate compound of formula (1a) is then treated with a compound of formula R 10 CO 2 H, where R 10 is not the same as R 10 present in the first tubulysin used to prepare the second compound of formula (2a).
  • Z is methyl.
  • R 1 is H.
  • R 1 is OR 6 at C(4), where R 6 is H, alkyl, or COR 7 .
  • V is H, and W is OC(O)R 3 .
  • R is OH.
  • R forms an ester derivative.
  • R forms an amide derivative.
  • R forms an acylhydrazide derivative, such as the compound formed from hydrazine.
  • a first tubulysin is converted into a second tubulysin by preparing an intermediate compound of formula (1b), wherein V is H, OR 2 , or halo, and W is H, OR 2 , or alkyl, where R 2 is independently selected in each instance from H, alkyl, or COR 3 , where R 3 is alkyl, alkenyl or aryl, providing that R 2 is not H when both V and W are OR 2 ; or V and W are taken together with the attached carbon to form a carbonyl; Z is CH 3 or COR 4 , and Y is absent; or Z is CH 3 and, Y is O; where R 4 is alkyl, CF 3 or aryl; T is H or OR 6 , where R 6 is H, alkyl, aryl, COR 7 , P(O)(OR 8 ) 2 , or SO 3 R 8 , where R 7 and R 8 are independently selected in each instance
  • R 10 is H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted;
  • V is H, OR 2 , or halo, and W is H, OR 2 , or alkyl, where R 2 is independently selected in each instance from H, alkyl, or COR 3 , where R 3 is alkyl, alkenyl, cycloalkyl, aryl or arylalkyl, each of which is optionally substituted, providing that R 2 is not H when both V and W are OR 2 ; or V and W are taken together with the attached carbon to form a carbonyl;
  • Z is CH 3 or COR 4 , and Y is absent; or Z is CH 3 and, Y is O; where R 4 is alkyl, CF 3 or aryl;
  • T is H or OR 6 , where R 6 is H, alkyl, aryl, COR 7 , P(O)(OR 8 ) 2
  • a first tubulysin or a mixture of tubulysins is converted into a second tubulysin by preparing an intermediate compound of formula (1c), wherein T is H or OH.
  • the intermediate is prepared by mixing a tubulysin or mixture of tubulysins of formula (2c) with an acid under substantially anhydrous conditions:
  • T is H or OH and R 10 is H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
  • R 10 is then treated with a compound of formula R 10 CO 2 H where R 10 is not the same as R 1 present in the starting tubulysin used to prepare the second compound of formula (2c).
  • tubulysins may instead be converted into different mixtures of tubulysins rather than a single tubulysin.
  • a single starting tubulysin, or mixture of tubulysins may be first converted into a common intermediate compound or a mixture of intermediate compounds of formulae (1a), (1b), or (1c), then those intermediate compounds are reacted with a mixture of carboxylic acids to provide the desired mixture of tubulysins.
  • tubulysin A a single starting tubulysin, such as tubulysin A, or the corresponding analog or derivative thereof, or alternatively a mixture of tubulysins
  • a single starting tubulysin such as tubulysin A, or the corresponding analog or derivative thereof, or alternatively a mixture of tubulysins
  • T is OH
  • carboxylic acids such as butanoic acid and propanoic acid
  • intermediate compound of formula (1b) can be treated with a compound of formula R 9 QH or the anion prepared therefrom to give a compound of the following formula:
  • R 9 is H, alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted, or R 9 is C(O)R 20 , S(O) 2 R 20 , or P(O)(OR 20 ) 2 ; where R 20 is independently selected in each instance from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted, or R 20 is a metal cation; V is H, OR 2 , or halo, and W is H, OR 2 , or alkyl, where R 2 is independently selected in each instance from H, alkyl, or COR 3 , where R 3 is alkyl, alkenyl, cycloalkyl, aryl or arylalkyl, each of which is optionally substituted, providing that R 2 is
  • intermediate compound of formula (1c) can be treated with a compound of formula R 9 QH or an anion thereof to give a compound of the following formula:
  • R 9 is H, alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted, or R 9 is C(O)R 20 , S(O) 2 R 20 , or P(O)(OR 20 ) 2 ; where R 20 is independently selected in each instance from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted, or R 20 is a metal cation: and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
  • intermediate compound of formula (1b) can be treated with a nitrile compound, R 21 CN, to give a compound of the following formula:
  • R 21 is alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted;
  • intermediate compound of formula (1c) can be treated with a nitrile compound, R 21 CN, to give a compound of the following formula:
  • R 21 is alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
  • intermediate compound of formula (1b) can be treated with an alkenylsilane of formula R 22 (TMS)CCH 2 to give a compound of the following formula:
  • R 22 is alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted;
  • V is H, OR 2 , or halo, and W is H, OR 2 , or alkyl, where R 2 is independently selected in each instance from H, alkyl, or COR 3 , where R 3 is alkyl, alkenyl, cycloalkyl, aryl or arylalkyl, each of which is optionally substituted, providing that R 2 is not H when both V and W are OR 2 ; or V and W are taken together with the attached carbon to form a carbonyl;
  • Z is CH 3 or COR 4 , and Y is absent; or Z is CH 3 and, Y is O; where R 4 is alkyl, CF 3 or aryl;
  • T is H or OR 6 , where R 6 is H, alkyl, aryl, COR 7 , P(O)(OR 8 ) 2 ,
  • intermediate compound of formula (1c) can be treated with an alkenylsilane of formula R 22 (TMS)CCH 2 to give a compound of the following formula:
  • R 22 is alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted; and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
  • olefins may form by isomerization, depending on the conditions of the reaction and the identity of R 22 .
  • R 22 is alkyl
  • the double bond can migrate to other carbon atoms along the alkenyl chain, including to form the terminal or ⁇ -olefin.
  • intermediate compound of formula (1b) is treated with a compound of formula R 23 C(O)CH 2 R a to give a compound of the following formula:
  • R 23 is H, alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted;
  • R a is C(O)R 9 , C(O)OR 9 or CN;
  • R 9 is selected from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted;
  • V is H, OR 2 , or halo
  • W is H, OR 2 , or alkyl, where R 2 is independently selected in each instance from H, alkyl, or COR 3 , where R 3 is alkyl, alkenyl, cycloalkyl, aryl or arylalkyl, each of which is optionally substituted, providing that R 2 is not H when both V and W are OR 2 ; or V and W are taken together with the attached carbon to form a carbonyl;
  • Z is CH 3 or COR 4
  • intermediate compound of formula (1c) is treated with a compound of formula R 23 C(O)CH 2 R a to give a compound of the following formula:
  • R 23 is H, alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted;
  • R a is C(O)R 9 , C(O)OR 9 or CN;
  • R 9 is selected from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted: and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
  • intermediate compound of formula (1b) is treated with water to give a second intermediate compound of the following formula (3b)
  • V is H, OR 2 , or halo
  • W is H, OR 2 , or alkyl
  • R 2 is independently selected in each instance from H, alkyl, or COR 3 , where R 3 is alkyl, alkenyl, cycloalkyl, aryl or arylalkyl, each of which is optionally substituted, providing that R 2 is not H when both V and W are OR 2 ; or V and W are taken together with the attached carbon to form a carbonyl
  • Z is CH 3 or COR 4 , and Y is absent; or Z is CH 3 and, Y is O; where R 4 is alkyl, CF 3 or aryl
  • T is H or OR 6 , where R 6 is H, alkyl, aryl, COR 7 , P(O)(OR 8 ) 2 , or SO 3 R 8 , where R 7 and R 8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl,
  • intermediate compound of formula (1c) is treated with water to give a second intermediate compound of formula (3c):
  • T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
  • intermediate compound of formula (3b) is treated with a halogenating, sulfonylating, phosphonylating or phosphorylation reagent to give a compound of the following formula:
  • X 3 is halogen, OS(O) 2 R 24 , OP(O)(OR 24 )R 24 , or OP(O)(OR 24 ) 2 ; where R 24 is independently selected in each instance from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted, or R 24 is a metal cation; V is H, OR 2 , or halo, and W is H, OR 2 , or alkyl, where R 2 is independently selected in each instance from H, alkyl, or COR 3 , where R 3 is alkyl, alkenyl, cycloalkyl, aryl or arylalkyl, each of which is optionally substituted, providing that R 2 is not H when both V and W are OR 2 ; or V and W are taken together with the attached carbon to form a carbonyl; Z is CH 3 or COR 4 , and Y is absent; or Z
  • intermediate compound of formula (3c) is treated with a halogenating, sulfonylating, phosphonylating or phosphorylation reagent to give a compound of the following formula:
  • X 3 is halogen, OS(O) 2 R 24 , OP(O)(OR 24 )R 24 , or OP(O)(OR 24 ) 2 ; where R 24 is independently selected in each instance from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted, or R 24 is a metal cation; and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
  • the corresponding compounds of formula (3a) may be similarly prepared via the corresponding intermediate of formula (1a) and treatment with a halogenating, sulfonylating, phosphonylating or phosphorylation reagent.
  • R 10 is H, alkyl, cycloalkyl, alkenyl, aryl or arylalkyl, each of which is optionally substituted: and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH; is treated with trifluoroacetic acid and the mixture is concentrated under reduced pressure to give an intermediate iminium compound.
  • iminium compounds may be present as the corresponding trifluoroacetate salt compounds, and other hydrates and solvates, and as the acylaminal compound, and other addition adducts, as is illustrated by the following formulae:
  • R 9 is H, alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted, or R 9 is C(O)R 20 , S(O) 2 R 20 , or P(O)(OR 20 ) 2 ; where R 20 is independently selected in each instance from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted, or R 20 is a metal cation: and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
  • R 9 is H, alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted, or R 9 is C(O)R 20 , S(O) 2 R 20 , or P(O)(OR 20 ) 2 ; where R 20 is independently selected in each instance from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted, or R 20 is a metal cation: and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
  • R 21 is alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted; and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
  • R 23 is H, alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted;
  • R a is C(O)R 8 , C(O)OR 8 or CN;
  • R 8 is selected from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted;
  • T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
  • R 22 is alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted; and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
  • hydroxytubulysin D when T is OH
  • hydroxytubulysin A when T is OH
  • R is other than OH
  • N-hydroxymethylubulysin A or N-hydroxymethylubulysin D is treated with a sulfonyl halide and a base to give a compound of the following formula:
  • R 24 is alkyl, cycloalkyl, alkenyl, aryl or arylalkyl, each of which is optionally substituted; and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
  • N-hydroxymethylubulysin A or N-hydroxymethylubulysin D is treated with a bromine or iodine in the presence of triphenylphospine and imidazole to give a compound of the following formula:
  • conjugates of tubulysins of the following formula are described:
  • T is H or OR 6 , where R 6 is H, alkyl, aryl, COR 7 , P(O)(OR 8 ) 2 , or SO 3 R 8 , where R 7 and R 8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R 8 is a metal cation, or R 6 is a phenol protecting group, or a prodrug moiety;
  • Z is alkyl or C(O)R 4 , where R 4 is alkyl, CF 3 , or aryl; and R is OH or a leaving group, or R forms a carboxylic acid derivative.
  • conjugates described herein may include spacer linkers and/or releasable linkers as generally described in US Patent Application Publication 2005/0002942, the disclosure of which is incorporated herein by reference.
  • conjugates described herein may include targeting ligands, including but not limited to folate and analogs and derivatives of folate, for targeting the conjugates to pathogenic cell populations, such as generally described in US Patent Application Publication 2005/0002942.
  • tubulysins of the following general formula are described
  • n 1-3;
  • V is H, OR 2 , or halo
  • W is H, OR 2 , or alkyl, where R 2 is independently selected in each instance from H, alkyl, or COR 3 , where R 3 is alkyl, alkenyl or aryl, providing that R 2 is not H when both V and W are OR 2 ; or V and W are taken together with the attached carbon to form a carbonyl;
  • Z is CH 3 or COR 4 , and Y is absent; or Z is CH 3 and, Y is O; where R 4 is alkyl, CF 3 or aryl;
  • T is H or OR 6 , where R 6 is H, alkyl, aryl, COR 7 , P(O)(OR 8 ) 2 , or SO 3 R 8 , where R 7 and R 8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R 8 is a metal cation;
  • S and U are each independently selected from the group consisting of H, halo, nitro, cyano, alkyl, haloalkyl, alkoxy, and haloalkoxy;
  • R is OH or a leaving group, or R forms a carboxylic acid derivative. Additional tubulysins are described in US patent application publication Nos. 2006/0128754 and 2005/0239713, the disclosures of which are incorporated herein by reference.
  • tubulysins of the following formula are described:
  • T is H or OR 6 , where R 6 is H, alkyl, aryl, COR 7 , P(O)(OR 8 ) 2 , or SO 3 R 8 , where R 7 and R 8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R 8 is a metal cation, or R 6 is a phenol protecting group, or a prodrug moiety;
  • Z is alkyl or C(O)R 4 , where R 4 is alkyl, CF 3 , or aryl; and R is OH or a leaving group, or R forms a carboxylic acid derivative.
  • tubulysins of the following formula are described:
  • n, S, T, U, V, W, Z, R, and R 10 are as described in the various embodiments herein.
  • tubulysins of the following formula are described:
  • n, S, T, U, V, W, Z, QR 9 , and R are as described in the various embodiments herein.
  • Q is —N—, —O—, or —S—; and R 9 is H, alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted.
  • QR 9 are taken together to form C(O)R 10 , S(O) 2 R 10 , P(O)(OR 10a ) 2 , where R 10 and OR 10a are independently selected in each instance from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted, or R 10a is a metal cation.
  • tubulysins of the following formula are described:
  • R 12 represents 1 or more substituents selected from alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted; and where n, S, T, U, V, W, Z, and R are as described in the various embodiments herein.
  • R 1 represents 1 or more substituents selected from alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted
  • n, S, T, U, V, W, Z, and R are as described in the various embodiments herein.
  • R 1 is alkyl
  • the double bond can migrate to other carbon atoms along the alkenyl chain, including to form the terminal or ⁇ -olefin.
  • tubulysins of the following formula are described:
  • R 13 is C(O)R 10 , C(O)OR 10 or CN; and where n, S, T, U, V, W, Z, R, and R 10 are as described in the various embodiments herein, where R 10 is independently selected in each instance.
  • tubulysins of the following formula are described:
  • n, S, T, U, V, W, Z, and R are as described in the various embodiments herein.
  • tubulysins of the following formula are described:
  • X 3 is halogen, OS(O) 2 R 10 , OP(O)(OR 10a )R 10 , or OP(O)(OR 10a ) 2 ; where R 10 and R 10a are independently selected in each instance from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted, or R 10a is a metal cation; and where n, S, T, U, V, W, Z, and R are as described in the various embodiments herein.
  • tubulysins useful in preparing the conjugates described herein are described in Peltier et al., “The Total Synthesis of Tubulysin D,” J. Am. Chem. Soc. 128:16018-19 (2006), the disclosure of which is incorporated herein by reference.
  • tubulysin compounds may be inhibitors of tubulin polymerization, and also may be DNA-alkylators. Accordingly, methods for treating diseases and disease states including pathogenic cell populations, such as cancer, are described herein.
  • Trifluoroacetic acid (TFA, 0.20 mL) was added via syringe into a light brown solution of a tubulysin mixture (20 mg, containing tubulysins A, B, C, G, I and hydroxytubulysin) in anhydrous dichloromethane (DCM, 0.80 mL).
  • the corresponding nucleophile for example, but not limited to, H 2 O, MeOH, 1-propanol, ethylene glycol, 3-methylbutanol, 1-propanethiol, 2-sulfanylethanol, 1,2-ethanedithiol, acetic acid, butyric acid, trans-4-chloro-2-butenoic acid
  • 0.20 mL was used for all thiols and 0.50 mL was used for all the others, and the solution was concentrated at reduced pressure on a Büchi Rotavapor and then further concentrated by means of an oil pump.
  • the crude product was dissolved in dimethyl sulfoxide (DMSO, 1.0 mL) and purified by preparative HPLC to afford the product as a white solid.
  • DMSO dimethyl sulfoxide
  • HPLC analysis indicated a complete conversion of the hydroxy tubulysin intermediate to tubulysin B.
  • the crude product was dissolved in DMSO (1.2 mL) and purified by preparative HPLC on a Waters XTerra Prep MS C 18 10 ⁇ m 19 ⁇ 250 mm column using a 25% B to 50% B gradient over 20 minutes (A: 2.0 mM phosphate buffer, pH 7.0; B: ACN) at 25 mL/minute. Fractions from 11.5 to 13.5 minutes were collected and lyophilized to 86 mg of a white solid containing 77 mg of tubulysin B and 9.0 mg of sodium phosphate salts.
  • tubulysin B As compared to a reference standard sample. It is to be understood that other mixtures of tubulysins can be similarly converted into a single tubulysin. It is further to be understood that this and other mixtures of tubulysins can be similarly converted into a different tubulysin than tubulysin B.
  • EXAMPLE Interconversion of natural tubulysins.
  • the conditions of the previous Example were repeated to (a) convert tubulysin A into tubulysin B, (b) convert tubulysin A into tubulysin I, (c) convert a mixture of tubulysins A and B into tubulysin B, and (d) convert a mixture of tubulysins A, B, and I into tubulysin B.
  • the yield of tubulysin B was ⁇ 90%. It is to be understood that in each of examples (a) to (d), the corresponding tubulysin can be converted into a different tubulysin than tubulysin B.
  • tubulysin B can be similarly converted into a different tubulysin.
  • N-hydroxymethyl substituted tubulysin was also isolated. It was surprisingly discovered that this hemiaminal was stable at neutral pH, and did not decompose to the free amine and formaldehyde.
  • R C(O)CH 2 CH(CH 3 ) 2 , C(O)CH 2 CH 2 CH 3 , C(O)CH 2 CH 3 , C(O)CH ⁇ C(CH 3 ) 2 , C(O)CH 3 , and H, respectively
  • TFA (0.15 mL) was added to a solution of the tubulysin mixture (19 mg) in anhydrous DCM (0.60 mL) at room temperature. After stirring for 40 minutes at room temperature under argon, the reaction was quenched with anhydrous MeOH (0.50 mL).
  • the solution was concentrated on a Büchi Rotavapor, co-evaporated with anhydrous MeOH (2 ⁇ ) and anhydrous DCM (2 ⁇ ), vacuumed for 30 minutes, co-evaporated again with anhydrous MeOH and anhydrous DCM (2 ⁇ ), and vacuumed for an additional 1.5 hours.
  • the residue was dissolved in anhydrous DCM (0.75 mL), to which was added allyltrimethylsilane (0.30 mL), cooled in an ice-bath, and to which was added BF 3 .Et 2 O (0.23 mL).
  • the reaction mixture was stirred under argon in an ice-bath for 30 minutes, and then the cooling was removed and the reaction mixture was stirred at room temperature for an additional 2 hours and 50 minutes.
  • N,N-Diisopropylethylamine (DIPEA, 6.1 ⁇ L) and isobutyl chloroformate (3.0 ⁇ L) were added with the help of a syringe in tandem into a solution of tubulysin B (0.15 mg) in anhydrous EtOAc (2.0 mL) at ⁇ 15° C.
  • EtOAc 2.0 mL
  • the reaction mixture was cooled down to ⁇ 20° C. and to which was added anhydrous hydrazine (5.0 ⁇ L).
  • the reaction mixture was stirred under argon at ⁇ 20° C.
  • additional tubulysin compounds and analogs and derivatives thereof.
  • additional ether forming alcohols may be used, including but not limited to alcohols, such as ethanol, propanol, sec-butanol, and the like, polyols, such as ethylene glycols, polyethylene glycols, propylene glycols, polypropylene glycols, glycerol, and the like, including alkyl, and acyl derivatives thereof, aminoalcohols, such as aminoethanol, aminopropanol, polyaminoalkylethanol, and the like, including alkyl, and acyl derivatives thereof, and others.
  • additional thiols, carboxylic acids, amino acids, amines, and the like may be used as nucleophiles to trap the intermediate iminium compounds of formulae (1) and (3).
  • METHOD EXAMPLE Inhibition of Cellular DNA Synthesis.
  • the compounds described herein are evaluated using an in vitro cytotoxicity assay that predicts the ability of the drug to inhibit the growth of folate receptor-positive KB cells.
  • the KB cells are exposed for up to 7 h at 37° C. over a range of concentrations of folate-drug conjugate in the absence or presence of at least a 100-fold excess of folic acid.
  • the cells are then rinsed once with fresh culture medium and incubated in fresh culture medium for 72 hours at 37° C. Cell viability is assessed using a 3 H-thymidine incorporation assay.
  • METHOD EXAMPLE In vitro concentration-dependent cytotoxic activity. Cells were heavily seeded in 24-well Falcon plates and allowed to form nearly confluent monolayers overnight. Thirty minutes prior to the addition of test article, spent medium was aspirated from all wells and replaced with fresh folate-free RPMI (FFRPMI). Note, designated wells received media containing 100 ⁇ M folic acid; and, cells within the latter wells were used to determine the targeting specificity, since cytotoxic activity produced in the presence of excess folic acid (enables competition for FR binding) would signify the portion of the total activity that was unrelated to FR-specific delivery.
  • FFRPMI folate-free RPMI
  • each well received 1 mL of media containing increasing concentrations of test article (4 wells per sample) in the presence or absence of 100 ⁇ M free folic acid (a binding site competitor).
  • Treated cells were pulsed for 2 h at 37° C., rinsed 4 times with 0.5 mL of media, and then chased in 1 mL of fresh media up to 70 h. Spent media was aspirated from all wells and replaced with fresh media containing 5 ⁇ Ci/mL 3 H-thymidine. Following a further 2 h 37° C.
  • IC 50 values concentration of drug conjugate required to reduce 3 H-thymidine incorporation into newly synthesized DNA by 50%
  • IC 50 values concentration of drug conjugate required to reduce 3 H-thymidine incorporation into newly synthesized DNA by 50%
  • the cytotoxicities of these conjugates are reduced in the presence of excess free folic acid, indicating that the observed cell killing was mediated by binding to the folate receptor.
  • mice Four to seven week-old mice (Balb/c or nu/nu strains) are purchased from Harlan Sprague Dawley, Inc. (Indianapolis, Ind.). Since normal rodent chow contains a high concentration of folic acid (6 mg/kg chow), mice used in these studies are maintained on the folate-free diet (Harlan diet #TD00434) for 1 week before tumor implantation to achieve serum folate concentrations close to the range of normal human serum. For tumor cell inoculation, 1 ⁇ 10 6 M109 cells or 1 ⁇ 10 6 KB cells in 100 ⁇ L are injected in the subcutis of the dorsal medial area.
  • Log cell kill (LCK) and treated over control (T/C) values are then calculated according to published procedures (see, e.g., Lee et al., “BMS-247550: a novel epothilone analog with a mode of action similar to paclitaxel but possessing superior antitumor efficacy” Clin Cancer Res 7:1429-1437 (2001); Rose, “Taxol-based combination chemotherapy and other in vivo preclinical antitumor studies” J Natl Cancer Inst Monogr 47-53 (1993)).
  • Dosing solutions are prepared fresh each day in PBS and administered through the lateral tail vein of the mice. Importantly, dosing is initiated when the s.c. tumors were between 50-100 mm 3 in volume.
  • Persistent drug toxicity is assessed by collecting blood via cardiac puncture and submitting the serum for independent analysis of blood urea nitrogen (BUN), creatinine, total protein, AST-SGOT, ALT-SGPT plus a standard hematological cell panel at Ani-Lytics, Inc. (Gaithersburg, Md.).
  • BUN blood urea nitrogen
  • AST-SGOT creatinine
  • ALT-SGPT ALT-SGPT plus a standard hematological cell panel at Ani-Lytics, Inc.
  • histopathologic evaluation of formalin-fixed heart, lungs, liver, spleen, kidney, intestine, skeletal muscle and bone is conducted by board-certified pathologists at Animal Reference Pathology Laboratories (ARUP; Salt Lake City, Utah).

Abstract

Processes are described for the preparation of tubulysins. The processes are useful for preparing predetermined mixtures of tubulysins, preparing single tubulysins from mixtures of tubulysins, and for converting one tubulysin into a different tubulysin. The tubulysins described herein are useful in treating diseases and disease states that include pathogenic cell populations.

Description

CROSS-REFERENCE TO RELATED APPLICATIONS
This application is a U.S. national application under 35 U.S.C. 371(b) of International Application Serial No. PCT/US2008/080948 filed Oct. 23, 2008, which claims the benefit under 35 U.S.C. §119(e) of U.S. Provisional Application Ser. No. 60/982,595, filed Oct. 25, 2007, and U.S. Provisional Application Ser. No. 61/036,176, filed Mar. 13, 2008, the disclosures of which are hereby incorporated herein by reference.
TECHNICAL FIELD
The invention described herein pertains to tubulysins and tubulysin analogs, and processes for preparing tubulysins and tubulysin analogs.
BACKGROUND
Tubulysins are a group of powerful inhibitors of tubulin polymerization. Tubulysins are useful in treating diseases and disease states that include pathogenic cell populations, such as cancer. Generally, tubulysins are linear tetrapeptides consisting of N-methyl pipecolic acid (Mep), isoleucine (Ile), an unnatural aminoacid called tubuvalin (Tuv), and either an unnatural aminoacid called tubutyrosine (Tut, an analog of tyrosine) or an unnatural aminoacid called tubuphenylalanine (Tup, an analog of phenylalanine), as shown in the following table:
Figure US09187521-20151117-C00001
Factor R1 T
A (CH3)2CHCH2 OH
B CH3(CH2)2 OH
C CH3CH2 OH
D (CH3)2CHCH2 H
E CH3(CH2)2 H
F CH2CH3 H
G (CH3)2C═CH OH
H CH3 H
I CH3 OH

Two particular species of mycobacteria synthesize tubulysins in high titer during fermentation. However, each species generally synthesizes a mixture of tubulysin factors, and that mixture differs between each of those mycobacteria species. For example, one species, Archangium gephyra, produces as the main component factors tubulysins A, B, C, G, and I, each of which may be identified by its including the Tut residue. In contrast, another species, Angiococcus disciformis, produces as the main component factors tubulysins D, E, F, and H, each of which may be identified by its including the Tup residue.
Such bacterial fermentations are convenient sources of tubulysins. However, because the mycobacteria produce only certain tubulysins, and/or mixtures of tubulysins, processes are needed for interconverting those tubulysins to the desired factors for medicinal and pharmacological uses. In addition, processes are needed for preparing novel tubulysins, tubulysin analogs, and tubulysin derivatives for medicinal and pharmacological uses.
SUMMARY OF THE INVENTION
Described herein are processes for preparing tubulysins. Also described herein are analogs and derivatives of tubulysins. In one embodiment, processes are described for preparing one or more tubulysins from a mixture of tubulysins, such as a mixture of tubulysins produced by fermentation or some other process. In another embodiment, processes are described herein for preparing a mixture of tubulysins from one or more tubulysins. In another embodiment, processes are described herein for converting one tubulysin into another tubulysin. In another embodiment, processes are described herein for converting one or more, or a mixture of tubulysins into one or more, or a mixture of tubulysin analogs. It is to be understood that as used herein, the term tubulysin refers both collectively and individually to the naturally occurring tubulysin, and the analogs and derivatives of tubulysins described herein, or that may be prepared from the processes described herein.
In another embodiment, novel tubulysins, tubulysin analogs, and tubulysin derivatives are described herein along with processes for preparing such novel tubulysins, tubulysin analogs, and tubulysin derivatives. In one embodiment, the processes include treating one or more tubulysins with an acid to prepare an intermediate. In another embodiment, the processes include the step of subsequently reacting the intermediate with a reagent to prepare a mixture of tubulysins from a different mixture of tubulysins, a single tubulysin from a mixture of tubulysins, a mixture of tubulysins from a single tubulysin, or a single tubulysins from a different tubulysin.
In one embodiment of the processes described herein, the intermediate is a compound of formulae (1a), (1b), or (1c):
Figure US09187521-20151117-C00002

wherein R, R1, S, T, U, V, W, Y, and Z are as described hereinbelow in the various embodiments, aspects, and variations thereof.
It is understood that intermediate compounds of formulae (1a), (1b), and (1c) may form salts, such as salts with the residual acid conjugate base. In addition, it is understood that iminium intermediates, such as those of formulae (1a), (1b), and (1c) may also be in equilibrium with the corresponding acyl aminal, where the residual acid conjugate base adds to the iminium intermediate. It is further appreciated that the iminium intermediates may form solvates or hydrates, each of which may be in equilibrium with the iminium intermediates. In any case, without being bound by theory, it is believed that nucleophiles such as RCN, RXH and the corresponding anions and salts thereof react with iminium intermediates regardless of whether the iminium is in a salt, hydrate, solvate or acylaminal form to prepare the compounds described herein, such as the compounds of formulae (2a), (2b), and (2c), respectively:
Figure US09187521-20151117-C00003

wherein R, R1, R10, S, T, U, V, W, Y, and Z are as described hereinbelow in the various embodiments, aspects, and variations thereof.
DETAILED DESCRIPTION
Described herein are processes for converting a single tubulysin or a mixture of tubulysin compounds into a single tubulysin compound or a different mixture of tubulysin compounds. In one embodiment, tubulysins described herein refer generally to tetrapeptide compounds of the formula
Figure US09187521-20151117-C00004

and pharmaceutical salts thereof, where
n is 1-3;
V is H, OR2, or halo, and W is H, OR2, or alkyl, where R2 is independently selected in each instance from H, alkyl, and C(O)R3, where R3 is alkyl, cycloalkyl, alkenyl, aryl, or arylalkyl, each of which is optionally substituted; providing that R2 is not H when both V and W are OR2; or V and W are taken together with the attached carbon to form a carbonyl;
X=H, C1-4 alkyl, alkenyl, each of which is optionally substituted, or CH2QR9; where Q is —N—, —O—, or —S—; R9=H, C1-4 alkyl, alkenyl, aryl, or C(O)R10; and R10=C1-6 alkyl, alkenyl, aryl, or heteroaryl;
Z is alkyl and Y is O; or Z is alkyl or C(O)R4, and Y is absent, where R4 is alkyl, CF3, or aryl;
R1 is H, or R1 represents 1 to 3 substituents selected from halo, nitro, carboxylate or a derivative thereof, cyano, hydroxyl, alkyl, haloalkyl, alkoxy, haloalkoxy, phenol protecting groups, prodrug moieties, and OR6, where R6 is optionally substituted aryl, C(O)R7, P(O)(OR8)2, or SO3R8, where R7 and R8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R8 is a metal cation; and
R is OH or a leaving group, or R forms a carboxylic acid derivative.
In one variation, Z is methyl. In another variation, R1 is H. In another variation, R1 is OR6 at C(4), where R6 is H, alkyl, or COR7. In another variation, V is H, and W is OC(O)R3.
Illustrative leaving groups include, but are not limited to halides, sulfonates, such as triflates, and the like, optionally substituted phenoxy, such as pentafluorophenoxy and the like, intermediates formed from ester forming or amide forming reagents, such as isobutyl chloroformate, DCC, HOBt, EDC, PyBOP, BOP, BOP-Cl, and the like.
Illustrative carboxylic acid derivatives include, but are not limited to, esters, amides, imides, acylhydrazides, nitriles, and optionally substituted variations thereof.
In another embodiment, tubulysins of the following general formula are described
Figure US09187521-20151117-C00005

and pharmaceutical salts thereof, where
n is 1-3;
V is H, OR2, or halo, and W is H, OR2, or alkyl, where R2 is independently selected in each instance from H, alkyl, or C(O)R3, where R3 is alkyl, alkenyl or aryl, providing that R2 is not H when both V and W are OR2; or V and W are taken together with the attached carbon to form a carbonyl;
X=H, C1-4 alkyl, alkenyl, each of which is optionally substituted, or CH2QR9; where Q is —N—, —O—, or —S—; R9=H, C1-4 alkyl, alkenyl, aryl, or C(O)R10; and R10=C1-6 alkyl, alkenyl, aryl, or heteroaryl;
Z is alkyl or C(O)R4, where R4 is alkyl, CF3, or aryl;
T is H or OR6, where R6 is H, alkyl, aryl, COR7, P(O)(OR8)2, or SO3R8, where R7 and R8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R8 is a metal cation, or R6 is a phenol protecting group, or a prodrug moiety;
S and U are each independently selected from the group consisting of H, halo, nitro, cyano, alkyl, haloalkyl, alkoxy, and haloalkoxy; and
R is OH or a leaving group, or R forms a carboxylic acid derivative.
In one embodiment, natural tubulysins, and the corresponding analogs and derivatives thereof are described. Such natural tubulysins are generally linear tetrapeptides consisting of N-methyl pipecolic acid (Mep), isoleucine (Ile), an unnatural aminoacid called tubuvalin (Tuv), and either an unnatural aminoacid called tubutyrosine (Tut, an analog of tyrosine) or an unnatural aminoacid called tubuphenylalanine (Tup, an analog of phenylalanine). In another embodiment, naturally occurring tubulysins, and analogs and derivatives thereof, of the following general formula are described
Figure US09187521-20151117-C00006

and pharmaceutical salts thereof, where R, R1, and R10 are as described in the various embodiments herein.
In one embodiment, a first tubulysin, or alternatively a mixture of tubulysins, is converted into a second tubulysin by preparing an intermediate compound of formula (1a), wherein
n is 1-3;
V is H, OR2, or halo, and W is H, OR2, or alkyl, where R2 is independently selected in each instance from H, alkyl, and C(O)R3, where R3 is alkyl, cycloalkyl, alkenyl, aryl, or arylalkyl, each of which is optionally substituted; providing that R2 is not H when both V and W are OR2; or V and W are taken together with the attached carbon to form a carbonyl;
X=H, C1-4 alkyl, alkenyl, each of which is optionally substituted, or CH2QR9; where Q is —N—, —O—, or —S—; R9=H, C1-4 alkyl, alkenyl, aryl, or C(O)R10; and R1=C1-6 alkyl, alkenyl, aryl, or heteroaryl;
Z is alkyl and Y is O; or Z is alkyl or C(O)R4, and Y is absent, where R4 is alkyl, CF3, or aryl;
R1 is H, or R1 represents 1 to 3 substituents selected from halo, nitro, carboxylate or a derivative thereof, cyano, hydroxyl, alkyl, haloalkyl, alkoxy, haloalkoxy, phenol protecting groups, prodrug moieties, and OR6, where R6 is optionally substituted aryl, C(O)R7, P(O)(OR8)2, or SO3R8, where R7 and R8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R8 is a metal cation; and
R is OH or a leaving group, or R forms a carboxylic acid derivative. The intermediate is prepared by mixing a tubulysin or mixture of tubulysins of formula (2b) with an acid under substantially anhydrous conditions:
Figure US09187521-20151117-C00007

and pharmaceutical salts thereof, where
n is 1-3;
V is H, OR2, or halo, and W is H, OR2, or alkyl, where R2 is independently selected in each instance from H, alkyl, and C(O)R3, where R3 is alkyl, cycloalkyl, alkenyl, aryl, or arylalkyl, each of which is optionally substituted; providing that R2 is not H when both V and W are OR2; or V and W are taken together with the attached carbon to form a carbonyl;
X=H, C1-4 alkyl, alkenyl, each of which is optionally substituted, or CH2QR9; where Q is —N—, —O—, or —S—; R9=H, C1-4 alkyl, alkenyl, aryl, or C(O)R10; and R10=C1-6 alkyl, alkenyl, aryl, or heteroaryl;
Z is alkyl and Y is O; or Z is alkyl or C(O)R4, and Y is absent, where R4 is alkyl, CF3, or aryl;
R1 is H, or R1 represents 1 to 3 substituents selected from halo, nitro, carboxylate or a derivative thereof, cyano, hydroxyl, alkyl, haloalkyl, alkoxy, haloalkoxy, phenol protecting groups, prodrug moieties, and OR6, where R6 is optionally substituted aryl, C(O)R7, P(O)(OR8)2, or SO3R8, where R7 and R8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R8 is a metal cation; and
R is OH or a leaving group, or R forms a carboxylic acid derivative.
The intermediate compound of formula (1a) is then treated with a compound of formula R10CO2H, where R10 is not the same as R10 present in the first tubulysin used to prepare the second compound of formula (2a).
In one variation, Z is methyl. In another variation, R1 is H. In another variation, R1 is OR6 at C(4), where R6 is H, alkyl, or COR7. In another variation, V is H, and W is OC(O)R3. In another variation, R is OH. In another variation, R forms an ester derivative. In another variation, R forms an amide derivative. In another variation, R forms an acylhydrazide derivative, such as the compound formed from hydrazine.
In another embodiment, a first tubulysin, or alternatively a mixture of tubulysins, is converted into a second tubulysin by preparing an intermediate compound of formula (1b), wherein V is H, OR2, or halo, and W is H, OR2, or alkyl, where R2 is independently selected in each instance from H, alkyl, or COR3, where R3 is alkyl, alkenyl or aryl, providing that R2 is not H when both V and W are OR2; or V and W are taken together with the attached carbon to form a carbonyl; Z is CH3 or COR4, and Y is absent; or Z is CH3 and, Y is O; where R4 is alkyl, CF3 or aryl; T is H or OR6, where R6 is H, alkyl, aryl, COR7, P(O)(OR8)2, or SO3R8, where R7 and R8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R8 is a metal cation; and S and U are each independently selected from the group consisting of H, halo, nitro, cyano, alkyl, haloalkyl, alkoxy, and haloalkoxy, or the corresponding carboxylic acid derivative thereof, where R is other than OH. The intermediate is prepared by mixing a tubulysin or mixture of tubulysins of formula (2b) with an acid under substantially anhydrous conditions:
Figure US09187521-20151117-C00008

wherein R10 is H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted; V is H, OR2, or halo, and W is H, OR2, or alkyl, where R2 is independently selected in each instance from H, alkyl, or COR3, where R3 is alkyl, alkenyl, cycloalkyl, aryl or arylalkyl, each of which is optionally substituted, providing that R2 is not H when both V and W are OR2; or V and W are taken together with the attached carbon to form a carbonyl; Z is CH3 or COR4, and Y is absent; or Z is CH3 and, Y is O; where R4 is alkyl, CF3 or aryl; T is H or OR6, where R6 is H, alkyl, aryl, COR7, P(O)(OR8)2, or SO3R8, where R7 and R8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R8 is a metal cation; and S and U are each independently selected from the group consisting of H, halo, nitro, cyano, alkyl, haloalkyl, alkoxy, and haloalkoxy, or the corresponding carboxylic acid derivative thereof, where R is other than OH. The intermediate compound of formula (1b) is then treated with a compound of formula R10CO2H, where R10 is not the same as R10 in the first tubulysin to prepare the second compound of formula (2b).
In another embodiment, a first tubulysin or a mixture of tubulysins is converted into a second tubulysin by preparing an intermediate compound of formula (1c), wherein T is H or OH. The intermediate is prepared by mixing a tubulysin or mixture of tubulysins of formula (2c) with an acid under substantially anhydrous conditions:
Figure US09187521-20151117-C00009

wherein T is H or OH and R10 is H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted, or the corresponding carboxylic acid derivative thereof, where R is other than OH. The intermediate compound of formula (1c) is then treated with a compound of formula R10CO2H where R10 is not the same as R1 present in the starting tubulysin used to prepare the second compound of formula (2c).
In each of the foregoing embodiments, it is understood that in certain configurations, the tubulysins may instead be converted into different mixtures of tubulysins rather than a single tubulysin. In such embodiments, a single starting tubulysin, or mixture of tubulysins, may be first converted into a common intermediate compound or a mixture of intermediate compounds of formulae (1a), (1b), or (1c), then those intermediate compounds are reacted with a mixture of carboxylic acids to provide the desired mixture of tubulysins. For example, if a mixture of tubulysins B and C, or the corresponding analogs or derivatives thereof, is desired, a single starting tubulysin, such as tubulysin A, or the corresponding analog or derivative thereof, or alternatively a mixture of tubulysins, may be first converted into the corresponding compound of formulae (1a), (1b), or (1c), wherein T is OH, then reacted with a mixture of carboxylic acids, such as butanoic acid and propanoic acid, corresponding to the appropriate groups on tubulysin B and C, respectively. It is appreciated that a statistical mixture may result based on the relative proportion of carboxylic acids used in the second step. Alternatively, it is also appreciated that kinetic and thermodynamic factors may influence the final proportion of tubulysins obtained in the second step and thus a statistical mixture of tubulysins based on the ratio of carboxylic acids will not result. In either case, it is understood that routine optimization of the relative ratios of carboxylic acid mixtures will afford the desired mixture of tubulysins from common intermediates of formulae (1a), (1b), and (1c).
In another embodiment, intermediate compound of formula (1b) can be treated with a compound of formula R9QH or the anion prepared therefrom to give a compound of the following formula:
Figure US09187521-20151117-C00010

wherein Q is —N—, —O—, or —S—; R9 is H, alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted, or R9 is C(O)R20, S(O)2R20, or P(O)(OR20)2; where R20 is independently selected in each instance from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted, or R20 is a metal cation; V is H, OR2, or halo, and W is H, OR2, or alkyl, where R2 is independently selected in each instance from H, alkyl, or COR3, where R3 is alkyl, alkenyl, cycloalkyl, aryl or arylalkyl, each of which is optionally substituted, providing that R2 is not H when both V and W are OR2; or V and W are taken together with the attached carbon to form a carbonyl Z is CH3 or COR4, and Y is absent; or Z is CH3 and, Y is O; where R4 is alkyl, CF3 or aryl; T is H or OR6, where R6 is H, alkyl, aryl, COR7, P(O)(OR8)2, or SO3R8, where R7 and R8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R8 is a metal cation; and S and U are each independently selected from the group consisting of H, halo, nitro, cyano, alkyl, haloalkyl, alkoxy, and haloalkoxy, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
In another embodiment, intermediate compound of formula (1c) can be treated with a compound of formula R9QH or an anion thereof to give a compound of the following formula:
Figure US09187521-20151117-C00011

wherein Q is —N—, —O—, or —S—; R9 is H, alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted, or R9 is C(O)R20, S(O)2R20, or P(O)(OR20)2; where R20 is independently selected in each instance from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted, or R20 is a metal cation: and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
It is to be understood that the corresponding compounds of formula (2a) may be similarly prepared via the corresponding intermediate of formula (1a) and R9QH.
In another embodiment, intermediate compound of formula (1b) can be treated with a nitrile compound, R21CN, to give a compound of the following formula:
Figure US09187521-20151117-C00012

wherein R21 is alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted; V is H, OR2, or halo, and W is H, OR2, or alkyl, where R2 is independently selected in each instance from H, alkyl, or COR3, where R3 is alkyl, alkenyl, cycloalkyl, aryl or arylalkyl, each of which is optionally substituted, providing that R2 is not H when both V and W are OR2; or V and W are taken together with the attached carbon to form a carbonyl Z is CH3 or COR4, and Y is absent; or Z is CH3 and, Y is O; where R4 is alkyl, CF3 or aryl; T is H or OR6, where R6 is H, alkyl, aryl, COR7, P(O)(OR8)2, or SO3R8, where R7 and R8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R8 is a metal cation; and S and U are each independently selected from the group consisting of H, halo, nitro, cyano, alkyl, haloalkyl, alkoxy, and haloalkoxy, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
In another embodiment, intermediate compound of formula (1c) can be treated with a nitrile compound, R21CN, to give a compound of the following formula:
Figure US09187521-20151117-C00013

wherein R21 is alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
It is to be understood that the corresponding compounds of formula (2a) may be similarly prepared via the corresponding intermediate of formula (1a) and R21CN.
In another embodiment, intermediate compound of formula (1b) can be treated with an alkenylsilane of formula R22(TMS)CCH2 to give a compound of the following formula:
Figure US09187521-20151117-C00014

wherein R22 is alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted; V is H, OR2, or halo, and W is H, OR2, or alkyl, where R2 is independently selected in each instance from H, alkyl, or COR3, where R3 is alkyl, alkenyl, cycloalkyl, aryl or arylalkyl, each of which is optionally substituted, providing that R2 is not H when both V and W are OR2; or V and W are taken together with the attached carbon to form a carbonyl; Z is CH3 or COR4, and Y is absent; or Z is CH3 and, Y is O; where R4 is alkyl, CF3 or aryl; T is H or OR6, where R6 is H, alkyl, aryl, COR7, P(O)(OR8)2, or SO3R8, where R7 and R8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R8 is a metal cation; and S and U are each independently selected from the group consisting of H, halo, nitro, cyano, alkyl, haloalkyl, alkoxy, and haloalkoxy, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
In another embodiment, intermediate compound of formula (1c) can be treated with an alkenylsilane of formula R22(TMS)CCH2 to give a compound of the following formula:
Figure US09187521-20151117-C00015

wherein R22 is alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted; and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
It is to be understood that the corresponding compounds of formula (2a) may be similarly prepared via the corresponding intermediate of formula (1a) and R22(TMS)CCH2.
It is understood that in the preceding embodiments other olefins may form by isomerization, depending on the conditions of the reaction and the identity of R22. For example, when R22 is alkyl, it is appreciated that under the reaction conditions, the double bond can migrate to other carbon atoms along the alkenyl chain, including to form the terminal or ω-olefin.
In another embodiment, intermediate compound of formula (1b) is treated with a compound of formula R23C(O)CH2Ra to give a compound of the following formula:
Figure US09187521-20151117-C00016

wherein R23 is H, alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted; Ra is C(O)R9, C(O)OR9 or CN; R9 is selected from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted; V is H, OR2, or halo, and W is H, OR2, or alkyl, where R2 is independently selected in each instance from H, alkyl, or COR3, where R3 is alkyl, alkenyl, cycloalkyl, aryl or arylalkyl, each of which is optionally substituted, providing that R2 is not H when both V and W are OR2; or V and W are taken together with the attached carbon to form a carbonyl; Z is CH3 or COR4, and Y is absent; or Z is CH3 and, Y is O; where R4 is alkyl, CF3 or aryl; T is H or OR6, where R6 is H, alkyl, aryl, COR7, P(O)(OR8)2, or SO3R8, where R7 and R8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R8 is a metal cation; and S and U are each independently selected from the group consisting of H, halo, nitro, cyano, alkyl, haloalkyl, alkoxy, and haloalkoxy, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
In another embodiment, intermediate compound of formula (1c) is treated with a compound of formula R23C(O)CH2Ra to give a compound of the following formula:
Figure US09187521-20151117-C00017

wherein R23 is H, alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted; Ra is C(O)R9, C(O)OR9 or CN; R9 is selected from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted: and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
It is to be understood that the corresponding compounds of formula (2a) may be similarly prepared via the corresponding intermediate of formula (1a) and R23C(O)CH2Ra.
In another embodiment, intermediate compound of formula (1b) is treated with water to give a second intermediate compound of the following formula (3b)
Figure US09187521-20151117-C00018

wherein V is H, OR2, or halo, and W is H, OR2, or alkyl, where R2 is independently selected in each instance from H, alkyl, or COR3, where R3 is alkyl, alkenyl, cycloalkyl, aryl or arylalkyl, each of which is optionally substituted, providing that R2 is not H when both V and W are OR2; or V and W are taken together with the attached carbon to form a carbonyl; Z is CH3 or COR4, and Y is absent; or Z is CH3 and, Y is O; where R4 is alkyl, CF3 or aryl; T is H or OR6, where R6 is H, alkyl, aryl, COR7, P(O)(OR8)2, or SO3R8, where R7 and R8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R8 is a metal cation; and S and U are each independently selected from the group consisting of H, halo, nitro, cyano, alkyl, haloalkyl, alkoxy, and haloalkoxy, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
In another embodiment, intermediate compound of formula (1c) is treated with water to give a second intermediate compound of formula (3c):
Figure US09187521-20151117-C00019

wherein T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
It is to be understood that the corresponding compounds of formula (3a) may be similarly prepared via the corresponding intermediate of formula (1a) and water.
In another embodiment, intermediate compound of formula (3b) is treated with a halogenating, sulfonylating, phosphonylating or phosphorylation reagent to give a compound of the following formula:
Figure US09187521-20151117-C00020

wherein X3 is halogen, OS(O)2R24, OP(O)(OR24)R24, or OP(O)(OR24)2; where R24 is independently selected in each instance from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted, or R24 is a metal cation; V is H, OR2, or halo, and W is H, OR2, or alkyl, where R2 is independently selected in each instance from H, alkyl, or COR3, where R3 is alkyl, alkenyl, cycloalkyl, aryl or arylalkyl, each of which is optionally substituted, providing that R2 is not H when both V and W are OR2; or V and W are taken together with the attached carbon to form a carbonyl; Z is CH3 or COR4, and Y is absent; or Z is CH3 and, Y is O; where R4 is alkyl, CF3 or aryl; T is H or OR6, where R6 is H, alkyl, aryl, COR7, P(O)(OR8)2, or SO3R8, where R7 and R8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R8 is a metal cation; and S and U are each independently selected from the group consisting of H, halo, nitro, cyano, alkyl, haloalkyl, alkoxy, and haloalkoxy, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
In another embodiment, intermediate compound of formula (3c) is treated with a halogenating, sulfonylating, phosphonylating or phosphorylation reagent to give a compound of the following formula:
Figure US09187521-20151117-C00021

wherein X3 is halogen, OS(O)2R24, OP(O)(OR24)R24, or OP(O)(OR24)2; where R24 is independently selected in each instance from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted, or R24 is a metal cation; and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
It is to be understood that the corresponding compounds of formula (3a) may be similarly prepared via the corresponding intermediate of formula (1a) and treatment with a halogenating, sulfonylating, phosphonylating or phosphorylation reagent.
In another embodiment, a compound of the following formula (2d):
Figure US09187521-20151117-C00022

is described, wherein R10 is H, alkyl, cycloalkyl, alkenyl, aryl or arylalkyl, each of which is optionally substituted: and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH; is treated with trifluoroacetic acid and the mixture is concentrated under reduced pressure to give an intermediate iminium compound. It is understood that such iminium compounds may be present as the corresponding trifluoroacetate salt compounds, and other hydrates and solvates, and as the acylaminal compound, and other addition adducts, as is illustrated by the following formulae:
Figure US09187521-20151117-C00023

Mixing the compound of those formulae, or other corresponding intermediates described herein and prepared by treatment of compound (2d) with TFA, and with an alcohol gives the corresponding N,O-acetal compound of the following formula:
Figure US09187521-20151117-C00024

wherein R9 is H, alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted, or R9 is C(O)R20, S(O)2R20, or P(O)(OR20)2; where R20 is independently selected in each instance from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted, or R20 is a metal cation: and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
In another embodiment, mixing a compound of formula (2d) with trifluoroacetic acid and concentrating the mixture under reduced pressure, followed by mixing the concentrated mixture with a thiol gives a N,S-acetal compound of the following formula:
Figure US09187521-20151117-C00025

wherein R9 is H, alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted, or R9 is C(O)R20, S(O)2R20, or P(O)(OR20)2; where R20 is independently selected in each instance from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted, or R20 is a metal cation: and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
In another embodiment, mixing a compound of formula (2d) with trifluoroacetic acid and concentrating the mixture under reduced pressure, followed by mixing the concentrated mixture with a nitrile gives a compound of the following formula:
Figure US09187521-20151117-C00026

wherein R21 is alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted; and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
In another embodiment, mixing a compound of formula (2d) with trifluoroacetic acid and concentrating the mixture under reduced pressure, followed by mixing the concentrated mixture with a ketone, in a Biginelli-type reaction, gives a compound of the following formula:
Figure US09187521-20151117-C00027

wherein R23 is H, alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted; Ra is C(O)R8, C(O)OR8 or CN; R8 is selected from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted; and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
In another embodiment, mixing a compound of formula (2d) with trifluoroacetic acid and concentrating the mixture under reduced pressure, followed by mixing the concentrated mixture with an alkenylsilane gives a compound of the following formula:
Figure US09187521-20151117-C00028

wherein R22 is alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted; and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
In another embodiment, mixing a compound of formula (2d) with trifluoroacetic acid and concentrating the mixture under reduced pressure, followed by mixing the concentrated mixture with water gives a compound of formula (3d):
Figure US09187521-20151117-C00029

which may also be referred to as hydroxytubulysin D when T is OH, or hydroxytubulysin A when T is OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
In another embodiment, N-hydroxymethylubulysin A or N-hydroxymethylubulysin D is treated with a sulfonyl halide and a base to give a compound of the following formula:
Figure US09187521-20151117-C00030

wherein R24 is alkyl, cycloalkyl, alkenyl, aryl or arylalkyl, each of which is optionally substituted; and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
In another embodiment, N-hydroxymethylubulysin A or N-hydroxymethylubulysin D is treated with a bromine or iodine in the presence of triphenylphospine and imidazole to give a compound of the following formula:
Figure US09187521-20151117-C00031

wherein X4 is Br or I; and T is H or OH, or the corresponding carboxylic acid derivative thereof, where R is other than OH.
In another embodiment, conjugates of tubulysins of the following formula are described:
Figure US09187521-20151117-C00032

and pharmaceutical salts thereof, where n is 1-3; T is H or OR6, where R6 is H, alkyl, aryl, COR7, P(O)(OR8)2, or SO3R8, where R7 and R8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R8 is a metal cation, or R6 is a phenol protecting group, or a prodrug moiety; Z is alkyl or C(O)R4, where R4 is alkyl, CF3, or aryl; and R is OH or a leaving group, or R forms a carboxylic acid derivative. Illustrative examples of such compounds, and their preparation are described in J. Med. Chem. 51, 1530-1533 (2008), the disclosure of which is incorporated herein by reference. It is understood that the conjugates may be formed at any heteroatom in the foregoing formula by removing the corresponding hydrogen or other group, including but not limited to conjugates formed by removing the hydrogen from R when R=OH, from T when T=OH, and the like. Conjugates described herein may include spacer linkers and/or releasable linkers as generally described in US Patent Application Publication 2005/0002942, the disclosure of which is incorporated herein by reference. In addition, conjugates described herein may include targeting ligands, including but not limited to folate and analogs and derivatives of folate, for targeting the conjugates to pathogenic cell populations, such as generally described in US Patent Application Publication 2005/0002942.
In another embodiment, tubulysins of the following general formula are described
Figure US09187521-20151117-C00033

and pharmaceutical salts thereof, where
n is 1-3;
V is H, OR2, or halo, and W is H, OR2, or alkyl, where R2 is independently selected in each instance from H, alkyl, or COR3, where R3 is alkyl, alkenyl or aryl, providing that R2 is not H when both V and W are OR2; or V and W are taken together with the attached carbon to form a carbonyl;
X=H, C1-4 alkyl, alkenyl, each of which is optionally substituted, or CH2QR9; where Q is —N—, —O—, or —S—; R9=H, C1-4 alkyl, alkenyl, aryl, or C(O)R10; and R10=C1-6 alkyl, alkenyl, aryl, or heteroaryl;
Z is CH3 or COR4, and Y is absent; or Z is CH3 and, Y is O; where R4 is alkyl, CF3 or aryl;
T is H or OR6, where R6 is H, alkyl, aryl, COR7, P(O)(OR8)2, or SO3R8, where R7 and R8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R8 is a metal cation;
S and U are each independently selected from the group consisting of H, halo, nitro, cyano, alkyl, haloalkyl, alkoxy, and haloalkoxy; and
R is OH or a leaving group, or R forms a carboxylic acid derivative. Additional tubulysins are described in US patent application publication Nos. 2006/0128754 and 2005/0239713, the disclosures of which are incorporated herein by reference.
In another embodiment, tubulysins of the following formula are described:
Figure US09187521-20151117-C00034

and pharmaceutical salts thereof, where n is 1-3; T is H or OR6, where R6 is H, alkyl, aryl, COR7, P(O)(OR8)2, or SO3R8, where R7 and R8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R8 is a metal cation, or R6 is a phenol protecting group, or a prodrug moiety; Z is alkyl or C(O)R4, where R4 is alkyl, CF3, or aryl; and R is OH or a leaving group, or R forms a carboxylic acid derivative. Illustrative examples of such compounds, and their preparation are described in J. Med. Chem. 51, 1530-1533 (2008), the disclosure of which is incorporated herein by reference.
In another embodiment, tubulysins of the following formula are described:
Figure US09187521-20151117-C00035

and pharmaceutical salts thereof, where n, S, T, U, V, W, Z, R, and R10 are as described in the various embodiments herein.
In another embodiment, tubulysins of the following formula are described:
Figure US09187521-20151117-C00036

and pharmaceutical salts thereof, where n, S, T, U, V, W, Z, QR9, and R are as described in the various embodiments herein. In one variation, Q is —N—, —O—, or —S—; and R9 is H, alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted. In another variation, QR9 are taken together to form C(O)R10, S(O)2R10, P(O)(OR10a)2, where R10 and OR10a are independently selected in each instance from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted, or R10a is a metal cation.
In another embodiment, tubulysins of the following formula are described:
Figure US09187521-20151117-C00037

and pharmaceutical salts thereof, where R12 represents 1 or more substituents selected from alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted; and where n, S, T, U, V, W, Z, and R are as described in the various embodiments herein. It is to be understood that other olefins may form by isomerization, depending on the conditions of the reaction and the identity of R1. For example, when R1 is alkyl, it is appreciated that under the reaction conditions, the double bond can migrate to other carbon atoms along the alkenyl chain, including to form the terminal or ω-olefin.
In another embodiment, tubulysins of the following formula are described:
Figure US09187521-20151117-C00038

and pharmaceutical salts thereof, where R13 is C(O)R10, C(O)OR10 or CN; and where n, S, T, U, V, W, Z, R, and R10 are as described in the various embodiments herein, where R10 is independently selected in each instance.
In another embodiment, tubulysins of the following formula are described:
Figure US09187521-20151117-C00039

and pharmaceutical salts thereof, where n, S, T, U, V, W, Z, and R are as described in the various embodiments herein.
In another embodiment, tubulysins of the following formula are described:
Figure US09187521-20151117-C00040

and pharmaceutical salts thereof, where X3 is halogen, OS(O)2R10, OP(O)(OR10a)R10, or OP(O)(OR10a)2; where R10 and R10a are independently selected in each instance from the group consisting of H, alkyl, alkenyl, cycloalkyl, aryl, and arylalkyl, each of which is optionally substituted, or R10a is a metal cation; and where n, S, T, U, V, W, Z, and R are as described in the various embodiments herein.
Additional tubulysins useful in preparing the conjugates described herein are described in Peltier et al., “The Total Synthesis of Tubulysin D,” J. Am. Chem. Soc. 128:16018-19 (2006), the disclosure of which is incorporated herein by reference.
In each of the foregoing embodiments, it is understood that in one variation, the compounds of the various formulae have the following absolute configuration:
Figure US09187521-20151117-C00041

at the indicated asymmetric backbone carbon atoms.
As described herein, the tubulysin compounds may be inhibitors of tubulin polymerization, and also may be DNA-alkylators. Accordingly, methods for treating diseases and disease states including pathogenic cell populations, such as cancer, are described herein.
EXAMPLES General Procedures
Trifluoroacetic acid (TFA, 0.20 mL) was added via syringe into a light brown solution of a tubulysin mixture (20 mg, containing tubulysins A, B, C, G, I and hydroxytubulysin) in anhydrous dichloromethane (DCM, 0.80 mL). After stirring for 40 minutes at room temperature and under argon, to the solution was added the corresponding nucleophile (for example, but not limited to, H2O, MeOH, 1-propanol, ethylene glycol, 3-methylbutanol, 1-propanethiol, 2-sulfanylethanol, 1,2-ethanedithiol, acetic acid, butyric acid, trans-4-chloro-2-butenoic acid), 0.20 mL was used for all thiols and 0.50 mL was used for all the others, and the solution was concentrated at reduced pressure on a Büchi Rotavapor and then further concentrated by means of an oil pump. The crude product was dissolved in dimethyl sulfoxide (DMSO, 1.0 mL) and purified by preparative HPLC to afford the product as a white solid.
GENERAL PREPARATIVE HPLC PARAMETERS: Column: Waters XTerra Prep MS C18 10 μm, 19×250 mm; Mobile phase A: 2.0 mM sodium phosphate buffer, pH 7.0; Mobile phase B: acetonitrile; Method: 10% B to 80% B over 30 minutes, flow rate=26 mL/min.
EXAMPLE. Interconversion of natural tubulysins. TFA (1.0 mL) was added to a light brown solution of a mixture of tubulysins (105 mg tubulysins A, B, C, G, I and hydroxy-tubulysin A) in dry DCM (5.0 mL) and the resulting light brown-greenish solution was stirred at room temperature under argon for 50 minutes. LC/MS indicated that the tubulysin mixture was converted to hydroxy-tubulysin. To the solution was added butyric acid (10 mL) and the solution was first concentrated by evaporation to a small volume to remove TFA and DCM, and then further concentrated to a thick oil under vacuum over about 1 hour. HPLC analysis indicated a complete conversion of the hydroxy tubulysin intermediate to tubulysin B. The crude product was dissolved in DMSO (1.2 mL) and purified by preparative HPLC on a Waters XTerra Prep MS C18 10 μm 19×250 mm column using a 25% B to 50% B gradient over 20 minutes (A: 2.0 mM phosphate buffer, pH 7.0; B: ACN) at 25 mL/minute. Fractions from 11.5 to 13.5 minutes were collected and lyophilized to 86 mg of a white solid containing 77 mg of tubulysin B and 9.0 mg of sodium phosphate salts. Monitoring the interconversion throughout the experiment by HPLC showed conversion only to tubulysin B, as compared to a reference standard sample. It is to be understood that other mixtures of tubulysins can be similarly converted into a single tubulysin. It is further to be understood that this and other mixtures of tubulysins can be similarly converted into a different tubulysin than tubulysin B.
EXAMPLE. Interconversion of natural tubulysins. The conditions of the previous Example were repeated to (a) convert tubulysin A into tubulysin B, (b) convert tubulysin A into tubulysin I, (c) convert a mixture of tubulysins A and B into tubulysin B, and (d) convert a mixture of tubulysins A, B, and I into tubulysin B. In each case, the yield of tubulysin B was ≧90%. It is to be understood that in each of examples (a) to (d), the corresponding tubulysin can be converted into a different tubulysin than tubulysin B. It is further to be understood that tubulysin B can be similarly converted into a different tubulysin. In some cases, N-hydroxymethyl substituted tubulysin was also isolated. It was surprisingly discovered that this hemiaminal was stable at neutral pH, and did not decompose to the free amine and formaldehyde.
Figure US09187521-20151117-C00042

The 1H NMR spectrum of hydroxytubulysin A was consistent with the structure; the MS had an m/z=760. The 1H NMR spectrum of tubulysin B obtained in this experiment was consistent with the structure; the MS had an m/z=830.
EXAMPLE. Methoxy Tubulysin (EC0313).
Figure US09187521-20151117-C00043
TFA (0.20 mL) was added to a solution of tubulysin B (21.0 mg) in dry DCM (0.80 mL) and the mixture was stirred at room temperature under argon for 1 hour. MeOH (1.0 mL) was added to the stirring solution and the solvent was evaporated after 1 h. The residue was purified by preparative HPLC on a Waters XTerra Prep MS C18 10 μm 19×250 mm column using a 20% B to 50% B gradient over 25 minutes (A: 1.0 mM phosphate buffer, pH 7.0; B: ACN) at 10 mL/minute. Fractions from 18.8 to 22.3 minutes were collected and lyophilized to 18.0 mg of a white solid containing 15.5 mg of title compound and 2.5 mg of sodium phosphate salts. The 1H NMR spectrum of methoxy tubulysin obtained in this experiment was consistent with the structure; the MS had an m/z=774.
EXAMPLE. 2-Hydroxyethoxy Tubulysin (EC0346).
Figure US09187521-20151117-C00044
TFA (75 μL) was added to a solution of tubulysin B (4.8 mg) in dry DCM (0.30 mL) and the mixture was stirred at room temperature under argon for 1 hour. Ethylene glycol (0.30 mL) was added to the stirring solution and the solvent was evaporated after 30 min. The residue was purified by a preparative HPLC on a Waters Phenomenex Luna C18 10 μm 4.6×250 mm column using a 10% B to 80% B gradient over 20 minutes (A: 1.0 mM phosphate buffer, pH 7.0; B: ACN) at 2.5 mL/minute. Fractions from 6.3 to 6.7 minutes were collected and lyophilized to 4.9 mg of a white solid containing 4.8 mg of title compound and 0.1 mg of sodium phosphate salts. The 1H NMR spectrum of 2-hydroxyethoxy tubulysin obtained in this experiment was consistent with the structure; the MS had an m/z=804.
EXAMPLE. 2-Mercaptoethylthio Tubulysin (EC0374).
Figure US09187521-20151117-C00045
TFA (60 μL) was added to a solution of tubulysin B (6.9 mg) in dry DCM (0.54 mL) and the mixture was stirred at room temperature under argon for 30 minutes. 1,2-Ethanedithiol (2.0 μL) was added to the stirring solution and the solvent was evaporated after 5 h. The residue was purified by preparative HPLC on a Waters XTerra Prep MS C18 10 μm 19×250 mm column using a 10% B to 80% B gradient over 20 minutes (A: 2.0 mM phosphate buffer, pH 7.0; B: ACN) at 25 mL/minute. Fractions from 9.5 to 10.7 minutes were collected and lyophilized to 7.7 mg of a white solid containing 3.4 mg of title compound and 4.3 mg of sodium phosphate salts. The 1H NMR spectrum of 2-mercaptoethylthio tubulysin obtained in this experiment was consistent with the structure; the MS had an m/z=836.
EXAMPLE. Iso-Butyrylamidotubulysin (EC0585)
Figure US09187521-20151117-C00046
A mixture of tubulysins containing factors A, B, C, G, I, and hydroxy tubulysin (R=C(O)CH2CH(CH3)2, C(O)CH2CH2CH3, C(O)CH2CH3, C(O)CH═C(CH3)2, C(O)CH3, and H, respectively) was converted to a single tubulysin. To a solution of the tubulysin mixture (25 mg) in isovaleronitrile (150 μL) was added a solution containing TFA (30 μL), concentrated H2SO4 (20 μL), and isovaleronitrile (150 μL). After stirring at room temperature for 22 hours, the reaction was quenched with 2.0 mM sodium phosphate buffer (pH=7.0, 15 mL) and injected into a preparative HPLC for purification. Column: Waters XTerra Prep MS C18 10 μm, 19×250 mm; Mobile phase A: 2.0 mM sodium phosphate buffer, pH 7.0; Mobile phase B: acetonitrile; Method: 10% B to 80% B over 30 minutes, flow rate=26 mL/min. Fractions from 17.22-18.36 minutes were collected and lyophilized to produce EC0585 as a white solid (16 mg). The 1H NMR spectrum of iso-butyrylamidotubulysin obtained in this experiment was consistent with the structure; the MS had an m/z=843.
EXAMPLE. N-Homoallyl Tubulysin EC0550
Figure US09187521-20151117-C00047
A mixture of tubulysins containing factors A, B, C, G, I, and hydroxy tubulysin (R=C(O)CH2CH(CH3)2, C(O)CH2CH2CH3, C(O)CH2CH3, C(O)CH═C(CH3)2, C(O)CH3, and H, respectively) was converted to a single tubulysin. TFA (0.15 mL) was added to a solution of the tubulysin mixture (19 mg) in anhydrous DCM (0.60 mL) at room temperature. After stirring for 40 minutes at room temperature under argon, the reaction was quenched with anhydrous MeOH (0.50 mL). The solution was concentrated on a Büchi Rotavapor, co-evaporated with anhydrous MeOH (2×) and anhydrous DCM (2×), vacuumed for 30 minutes, co-evaporated again with anhydrous MeOH and anhydrous DCM (2×), and vacuumed for an additional 1.5 hours. The residue was dissolved in anhydrous DCM (0.75 mL), to which was added allyltrimethylsilane (0.30 mL), cooled in an ice-bath, and to which was added BF3.Et2O (0.23 mL). The reaction mixture was stirred under argon in an ice-bath for 30 minutes, and then the cooling was removed and the reaction mixture was stirred at room temperature for an additional 2 hours and 50 minutes. The reaction mixture was concentrated and the residue was purified by a preparative HPLC. Column: Waters XTerra Prep MS C18 10 μm, 19×250 mm; Mobile phase A: 2.0 mM sodium phosphate buffer, pH 7.0; Mobile phase B: acetonitrile; Method: 15% B to 80% B over 30 minutes, flow rate=26 mL/min. Fractions from 14.41-15.17 minutes were collected and lyophilized to afford EC0550 as a white solid (10 mg). The 1H NMR spectrum of N-homoallyl tubulysin obtained in this experiment was consistent with the structure; the MS had an m/z=784.
EXAMPLE. Synthesis of Tubulysin B Hydrazide EC0347.
Figure US09187521-20151117-C00048
N,N-Diisopropylethylamine (DIPEA, 6.1 μL) and isobutyl chloroformate (3.0 μL) were added with the help of a syringe in tandem into a solution of tubulysin B (0.15 mg) in anhydrous EtOAc (2.0 mL) at −15° C. After stirring for 45 minutes at −15° C. under argon, the reaction mixture was cooled down to −20° C. and to which was added anhydrous hydrazine (5.0 μL). The reaction mixture was stirred under argon at −20° C. for 3 hours, quenched with 1.0 mM sodium phosphate buffer (pH 7.0, 1.0 mL), and injected into a preparative HPLC for purification. Column: Waters XTerra Prep MS C18 10 μm, 19×250 mm; Mobile phase A: 1.0 mM sodium phosphate buffer, pH 7.0; Mobile phase B: acetonitrile; Method: 10% B to 80% B over 20 minutes, flow rate=25 mL/min. Fractions from 15.14-15.54 minutes were collected and lyophilized to produce EC0347 as a white solid (2.7 mg). The 1H NMR spectrum of tubulysin B hydrazide obtained in this experiment was consistent with the structure; the MS had an m/z=844.
EXAMPLE. Synthesis of EC0311.
Figure US09187521-20151117-C00049
DIPEA (0.60 mL) was added to a suspension of HOBt-A (685 mg, 91%) in anhydrous DCM (5.0 mL) at 0° C., stirred under argon for 2 minutes, and to which was added anhydrous hydrazine (0.10 mL). The reaction mixture was stirred under argon at 0° C. for 10 minutes and room temperature for an additional 30 minutes, filtered, and the filtrate was purified by flash chromatography (silica gel, 2% MeOH in DCM) to afford EC0311 as a clear thick oil (371 mg), solidified upon standing.
EXAMPLE. Synthesis of EC0312.
Figure US09187521-20151117-C00050
DIPEA (36 μL) and isobutyl chloroformate (13 μL) were added with the help of a syringe in tandem into a solution of tubulysin B (82 mg) in anhydrous EtOAc (2.0 mL) at −15° C. After stirring for 45 minutes at −15° C. under argon, to the reaction mixture was added a solution of EC0311 in anhydrous EtOAc (1.0 mL). The resulting solution was stirred under argon at −15° C. for 15 minutes and room temperature for an additional 45 minutes, concentrated, and the residue was purified by flash chromatography (silica gel, 2 to 8% MeOH in DCM) to give EC0312 as a white solid (98 mg). The 1H NMR spectrum of EC0312 obtained in this experiment was consistent with the structure; the MS had an m/z=1057.
EXAMPLE. The following compounds were prepared according to the procedures described herein.
Figure US09187521-20151117-C00051
The 1H NMR spectrum of EC0575 obtained in this experiment was consistent with the structure; the MS had an m/z=862.
Figure US09187521-20151117-C00052
The 1H NMR spectrum of EC0560 obtained in this experiment was consistent with the structure; the MS had an m/z=820.
Figure US09187521-20151117-C00053
The 1H NMR spectrum of EC0356 obtained in this experiment was consistent with the structure; the MS had an m/z=817.
Figure US09187521-20151117-C00054
The 1H NMR spectrum of EC0611 obtained in this experiment was consistent with the structure; the MS had an m/z=802.
Figure US09187521-20151117-C00055
The 1H NMR spectrum of EC0623 obtained in this experiment was consistent with the structure; the MS had an m/z=818.
It is appreciated that the foregoing Examples are merely illustrative of the processes described herein and that many routine modifications may be made to prepare additional tubulysin compounds, and analogs and derivatives thereof. For example, additional ether forming alcohols may be used, including but not limited to alcohols, such as ethanol, propanol, sec-butanol, and the like, polyols, such as ethylene glycols, polyethylene glycols, propylene glycols, polypropylene glycols, glycerol, and the like, including alkyl, and acyl derivatives thereof, aminoalcohols, such as aminoethanol, aminopropanol, polyaminoalkylethanol, and the like, including alkyl, and acyl derivatives thereof, and others. Similarly, additional thiols, carboxylic acids, amino acids, amines, and the like may be used as nucleophiles to trap the intermediate iminium compounds of formulae (1) and (3).
METHOD EXAMPLE. Inhibition of Cellular DNA Synthesis. The compounds described herein are evaluated using an in vitro cytotoxicity assay that predicts the ability of the drug to inhibit the growth of folate receptor-positive KB cells. The KB cells are exposed for up to 7 h at 37° C. over a range of concentrations of folate-drug conjugate in the absence or presence of at least a 100-fold excess of folic acid. The cells are then rinsed once with fresh culture medium and incubated in fresh culture medium for 72 hours at 37° C. Cell viability is assessed using a 3H-thymidine incorporation assay.
METHOD EXAMPLE. In vitro concentration-dependent cytotoxic activity. Cells were heavily seeded in 24-well Falcon plates and allowed to form nearly confluent monolayers overnight. Thirty minutes prior to the addition of test article, spent medium was aspirated from all wells and replaced with fresh folate-free RPMI (FFRPMI). Note, designated wells received media containing 100 μM folic acid; and, cells within the latter wells were used to determine the targeting specificity, since cytotoxic activity produced in the presence of excess folic acid (enables competition for FR binding) would signify the portion of the total activity that was unrelated to FR-specific delivery. Following one rinse with 1 mL of fresh FFRPMI containing 10% heat-inactivated fetal calf serum, each well received 1 mL of media containing increasing concentrations of test article (4 wells per sample) in the presence or absence of 100 μM free folic acid (a binding site competitor). Treated cells were pulsed for 2 h at 37° C., rinsed 4 times with 0.5 mL of media, and then chased in 1 mL of fresh media up to 70 h. Spent media was aspirated from all wells and replaced with fresh media containing 5 μCi/mL 3H-thymidine. Following a further 2 h 37° C. incubation, cells were washed 3 times with 0.5 mL of PBS and then treated with 0.5 mL of ice-cold 5% trichloroacetic acid per well. After 15 min, the trichloroacetic acid was aspirated and the cell material solubilized by the addition of 0.5 mL of 0.25 N sodium hydroxide for 15 min. Four hundred and fifty μL of each solubilized sample were transferred to scintillation vials containing 3 mL of Ecolume scintillation cocktail and then counted in a liquid scintillation counter. Final results are expressed as the percentage of 3H-thymidine incorporation relative to untreated controls.
As shown in the following table, dose-dependent cytotoxicity is measurable, and in most cases, the IC50 values (concentration of drug conjugate required to reduce 3H-thymidine incorporation into newly synthesized DNA by 50%) are in the low nanomolar range. Furthermore, the cytotoxicities of these conjugates are reduced in the presence of excess free folic acid, indicating that the observed cell killing was mediated by binding to the folate receptor.
Example Cytotoxicity IC50 (nM)
Tubulysin A 2
Tubulysin B 2.6
EC0313 9
EC0346 18
EC0550 2
EC0356 12
EC0374 24
EC0585 8
EC0386 42
EC0623 0.48
EC0346 18
METHOD EXAMPLE. Tumor models and therapy. Four to seven week-old mice (Balb/c or nu/nu strains) are purchased from Harlan Sprague Dawley, Inc. (Indianapolis, Ind.). Since normal rodent chow contains a high concentration of folic acid (6 mg/kg chow), mice used in these studies are maintained on the folate-free diet (Harlan diet #TD00434) for 1 week before tumor implantation to achieve serum folate concentrations close to the range of normal human serum. For tumor cell inoculation, 1×106 M109 cells or 1×106 KB cells in 100 μL are injected in the subcutis of the dorsal medial area. Tumors are measured in two perpendicular directions every 2-3 days using a caliper, and their volumes were calculated as 0.5×L×W2, where L=measurement of longest axis in mm and W=measurement of axis perpendicular to L in mm. Log cell kill (LCK) and treated over control (T/C) values are then calculated according to published procedures (see, e.g., Lee et al., “BMS-247550: a novel epothilone analog with a mode of action similar to paclitaxel but possessing superior antitumor efficacy” Clin Cancer Res 7:1429-1437 (2001); Rose, “Taxol-based combination chemotherapy and other in vivo preclinical antitumor studies” J Natl Cancer Inst Monogr 47-53 (1993)). Dosing solutions are prepared fresh each day in PBS and administered through the lateral tail vein of the mice. Importantly, dosing is initiated when the s.c. tumors were between 50-100 mm3 in volume.
Persistent drug toxicity is assessed by collecting blood via cardiac puncture and submitting the serum for independent analysis of blood urea nitrogen (BUN), creatinine, total protein, AST-SGOT, ALT-SGPT plus a standard hematological cell panel at Ani-Lytics, Inc. (Gaithersburg, Md.). In addition, histopathologic evaluation of formalin-fixed heart, lungs, liver, spleen, kidney, intestine, skeletal muscle and bone (tibia/fibula) is conducted by board-certified pathologists at Animal Reference Pathology Laboratories (ARUP; Salt Lake City, Utah).

Claims (29)

What is claimed is:
1. A process for converting one or more compounds of the formula I
Figure US09187521-20151117-C00056
wherein:
n is 1-3;
V is H, OR2, or halo, and W is H, OR2, or alkyl, where R2 is independently selected in each instance from H, alkyl, and C(O)R5, where R5 is independently selected in each instance from the group consisting of alkyl, cycloalkyl, alkenyl, arylalkyl and aryl; providing that R2 is not H when both V and W are OR2; or V and W are taken together with the attached carbon to form a carbonyl;
Z is alkyl and Y is O; or Z is alkyl or C(O)R4, and Y is absent, where R4 is alkyl, CF3, or aryl;
R1 is H, or R1 represents 1 to 3 substituents independently selected in each instance from the group consisting of halo, nitro, carboxylate or a derivative thereof, cyano, hydroxyl, alkyl, haloalkyl, alkoxy, haloalkoxy, and OR6, where R6 is an optionally substituted aryl, C(O)R7, P(O)(OR8)2, SO3R8, a phenol protecting group, or a prodrug moiety, where R7 and R8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R8 is a metal cation;
R is OH or a leaving group, or R forms a carboxylic acid derivative; and
R10 is independently selected in each instance from the group consisting of alkyl and alkenyl, each of which is optionally substituted; and where if there is a plurality of compounds of formula I, the plurality of compounds differ from each other in R10;
into substantially a single compound of the formula II
Figure US09187521-20151117-C00057
wherein
n, V, W, Z, Y, R1 and R are as defined in Formula I; and
X is CH2QR9; where Q is —N(R9)— or —S—, and each R9 is independently hydrogen, C1-4 alkyl, alkenyl, aryl, each of which is optionally substituted, or C(O)R11, where R11 is C1-6 alkyl, alkenyl, aryl, or heteroaryl, each of which is optionally substituted; or Q is —O—, and R9 is C(O)R11 or C5alkyl;
the process comprising the steps of (a) mixing the one or more of compounds of formula I with an acid under substantially anhydrous conditions to form an intermediate; and
(b) reacting the intermediate with a nucleophile.
2. The process of claim 1 for converting substantially one compound of formula I.
3. The process of claim 1 for converting a plurality of compounds of formula I.
4. The process of claim 1 wherein n is 2 and R is OH.
5. The process of claim 1 wherein V is H and W is OR2.
6. The process of claim 4 wherein the nucleophile is a compound of the formula R11CO2H.
7. The process of claim 4 wherein the nucleophile is a compound of formula R9QH or an anion thereof.
8. The process of claim 4 wherein the nucleophile is a compound of the formula R21CN, wherein R21 is alkyl, alkenyl, cycloalkyl, aryl, or arylalkyl, each of which is optionally substituted.
9. The process of claim 1 wherein each Z is alkyl; and each Y is absent.
10. The process of claim 1 wherein each n is 1.
11. The process of claim 1 wherein each n is 2.
12. The process of claim 1 wherein each R10 is alkyl.
13. The process of claim 1 wherein each V is H; and each W is OR2; where R2 is C(O)R5.
14. The process of claim 13 wherein R5 is alkyl.
15. The process of claim 1 wherein each R1 is H.
16. The process of claim 1 wherein each R1 is OH.
17. The process of claim 1 wherein each R is OH.
18. The process of claim 1 wherein Q is —O—; and R9 is C(O)R11.
19. The process of claim 18 wherein R11 is C1-6 alkyl.
20. The process of claim 18 wherein R11 is C1-6 alkenyl.
21. The process of claim 1 wherein Q is —NH—.
22. The process of claim 1 wherein Q is —S—.
23. The process of claim 1 wherein the one or more compounds of the formula I are of the formula.
Figure US09187521-20151117-C00058
24. The process of claim 1 wherein the compound of the formula II is of the formula.
Figure US09187521-20151117-C00059
25. The process of claim 1 wherein the one or more compounds of the formula I are of the formula
Figure US09187521-20151117-C00060
wherein R1 is OH and R10 is (CH3)2CHCH2; R1 is OH and R10 is CH3(CH2)2; R1 is OH and R10 is CH3CH2; R1 is H and R10 is (CH3)2CHCH2; R1 is H and R10 is CH3(CH2)2; R1 is H and R10 is CH2CH3; R1 is OH and R10 is (CH3)2C=CH; R1 is H and R10 is CH3; or R1 is H and R10 is CH3.
26. The process of claim 1 wherein the compound of the formula II is of the formula
Figure US09187521-20151117-C00061
wherein R1 is OH and R11 is (CH3)2CHCH2; R1 is OH and R11 is CH3(CH2)2; R1 is OH and R11 is CH3CH2; R1 is H and R11 is (CH3)2CHCH2; R1 is H and R11 is CH3(CH2)2; R1 is H and R11 is CH2CH3; R1 is OH and R11 is (CH3)2C=CH; R1 is H and R11 is CH3; or R1 is H and R11 is CH3.
27. The process of claim 1 wherein the compound of the formula II is tubulysin B.
28. The process of claim 1 wherein the compound of the formula II is selected from the group consisting of
Figure US09187521-20151117-C00062
29. A process for converting one or more compounds of the formula III
Figure US09187521-20151117-C00063
wherein:
n is 1-3;
V is H, OR2, or halo, and W is H, OR2, or alkyl, where R2 is independently selected in each instance from H, alkyl, and C(O)R5, where R5 is independently selected in each instance from the group consisting of alkyl, cycloalkyl, alkenyl, arylalkyl and aryl; providing that R2 is not H when both V and W are OR2; or V and W are taken together with the attached carbon to form a carbonyl;
Z is alkyl and Y is O; or Z is alkyl or C(O)R4, and Y is absent, where R4 is alkyl, CF3, or aryl;
R1 is H, or R1 represents 1 to 3 substituents independently selected in each instance from the group consisting of halo, nitro, carboxylate or a derivative thereof, cyano, hydroxyl, alkyl, haloalkyl, alkoxy, haloalkoxy, and OR6, where R6 is an optionally substituted aryl, C(O)R7, P(O)(OR8)2, SO3R8, a phenol protecting group, or a prodrug moiety, where R7 and R8 are independently selected in each instance from H, alkyl, alkenyl, cycloalkyl, heterocyclyl, aryl, and arylalkyl, each of which is optionally substituted, or R8 is a metal cation;
R is OH or a leaving group, or R forms a carboxylic acid derivative; and
R10 is independently selected in each instance from the group consisting of alkyl and alkenyl, each of which is optionally substituted; and where if there is a plurality of compounds of formula I, the plurality of compounds differ from each other in R10;
into substantially a single compound of the formula IV
Figure US09187521-20151117-C00064
wherein
n, V, W, Z, Y, R1 and R are as defined in Formula I; and
X is CH2QR9; where Q is -N(R9)- or -S-, and each R9 is independently hydrogen, C1-4 alkyl, alkenyl, aryl, each of which is optionally substituted, or C(O)R11, where R11 is C1-6 alkyl, alkenyl, aryl, or heteroaryl, each of which is optionally substituted; or Q is -O-, and R9 is C(O)R11;
the process comprising the steps of (a) mixing the one or more of compounds of formula I with an acid under substantially anhydrous conditions to form an iminium intermediate; and
(b) reacting the intermediate with a nucleophile.
US12/739,579 2007-10-25 2008-10-23 Tubulysins and processes for preparing Expired - Fee Related US9187521B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/739,579 US9187521B2 (en) 2007-10-25 2008-10-23 Tubulysins and processes for preparing

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US98259507P 2007-10-25 2007-10-25
US3617608P 2008-03-13 2008-03-13
PCT/US2008/080948 WO2009055562A1 (en) 2007-10-25 2008-10-23 Tubulysins and processes for preparing
US12/739,579 US9187521B2 (en) 2007-10-25 2008-10-23 Tubulysins and processes for preparing

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2008/080948 A-371-Of-International WO2009055562A1 (en) 2007-10-25 2008-10-23 Tubulysins and processes for preparing

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/921,196 Continuation US9745341B2 (en) 2007-10-25 2015-10-23 Tubulysins and processes for preparing

Publications (2)

Publication Number Publication Date
US20100240701A1 US20100240701A1 (en) 2010-09-23
US9187521B2 true US9187521B2 (en) 2015-11-17

Family

ID=40580005

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/739,579 Expired - Fee Related US9187521B2 (en) 2007-10-25 2008-10-23 Tubulysins and processes for preparing
US14/921,196 Expired - Fee Related US9745341B2 (en) 2007-10-25 2015-10-23 Tubulysins and processes for preparing

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/921,196 Expired - Fee Related US9745341B2 (en) 2007-10-25 2015-10-23 Tubulysins and processes for preparing

Country Status (8)

Country Link
US (2) US9187521B2 (en)
EP (1) EP2209374B1 (en)
JP (3) JP2011500835A (en)
CN (1) CN101909441B (en)
AU (1) AU2008316835B2 (en)
CA (1) CA2703491C (en)
IL (1) IL205253A (en)
WO (1) WO2009055562A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160016993A1 (en) * 2013-03-01 2016-01-21 Endocyte, Inc. Process for preparing tubulysins
US9745341B2 (en) 2007-10-25 2017-08-29 Endocyte, Inc. Tubulysins and processes for preparing

Families Citing this family (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006012527A1 (en) 2004-07-23 2006-02-02 Endocyte, Inc. Bivalent linkers and conjugates thereof
WO2008101231A2 (en) 2007-02-16 2008-08-21 Endocyte, Inc. Methods and compositions for treating and diagnosing kidney disease
EP2489372A3 (en) 2007-03-14 2013-01-02 Endocyte, Inc. Binding ligand linked drug delivery conjugates of tubulysins
EP2176293B1 (en) 2007-06-25 2019-04-03 Endocyte, Inc. Conjugates containing hydrophilic spacer linkers
US9877965B2 (en) 2007-06-25 2018-01-30 Endocyte, Inc. Vitamin receptor drug delivery conjugates for treating inflammation
EP2187965B1 (en) 2007-08-17 2019-10-09 Purdue Research Foundation Psma binding ligand-linker conjugates and methods for using
IT1394860B1 (en) * 2009-07-22 2012-07-20 Kemotech S R L PHARMACEUTICAL COMPOUNDS
US8394922B2 (en) 2009-08-03 2013-03-12 Medarex, Inc. Antiproliferative compounds, conjugates thereof, methods therefor, and uses thereof
CN102648208B (en) * 2009-11-12 2016-04-27 R&D生技药品有限责任公司 Antitubulin
US9951324B2 (en) 2010-02-25 2018-04-24 Purdue Research Foundation PSMA binding ligand-linker conjugates and methods for using
EP2409983A1 (en) * 2010-07-19 2012-01-25 Leibniz-Institut für Pflanzenbiochemie (IPB) Tubulysin analogues
EP2717916B1 (en) 2011-06-10 2017-03-08 Mersana Therapeutics, Inc. Protein-polymer-drug conjugates
US10080805B2 (en) 2012-02-24 2018-09-25 Purdue Research Foundation Cholecystokinin B receptor targeting for imaging and therapy
US20140080175A1 (en) * 2012-03-29 2014-03-20 Endocyte, Inc. Processes for preparing tubulysin derivatives and conjugates thereof
CA3102268C (en) 2012-07-12 2023-03-07 Hangzhou Dac Biotech Co., Ltd Conjugates of cell binding molecules with cytotoxic agents
BR112015008365A2 (en) 2012-10-16 2017-07-04 Endocyte Inc compound of the formula bl (d) x, or a pharmaceutically acceptable salt thereof, pharmaceutical composition, use of a compound, unit dosage form or unit dose composition, composition for treating a cancer in a patient, and method for treating a cancer in a patient
CA2891476C (en) 2012-11-15 2022-07-05 Endocyte, Inc. Drug delivery conjugates, and methods for treating diseases caused by psma expressing cells
JP6133431B2 (en) 2012-11-24 2017-05-24 ハンジョウ ディーエーシー バイオテック シーオー.,エルティディ.Hangzhou Dac Biotech Co.,Ltd. Use of hydrophilic conjugates and conjugation reactions between drug molecules and cell binding molecules
JP6334553B2 (en) 2012-12-10 2018-05-30 メルサナ セラピューティクス,インコーポレイティド Protein-polymer-drug conjugate
EP2931316B1 (en) 2012-12-12 2019-02-20 Mersana Therapeutics, Inc. Hydroxyl-polymer-drug-protein conjugates
KR102215954B1 (en) 2013-02-14 2021-02-15 브리스톨-마이어스 스큅 컴퍼니 Tubulysin compounds, methods of making and use
EP3038840B1 (en) * 2013-08-30 2022-03-16 Mubea Carbo Tech GmbH Composite wheel and insert
EP3415489A1 (en) 2013-10-18 2018-12-19 Deutsches Krebsforschungszentrum Labeled inhibitors of prostate specific membrane antigen (psma), their use as imaging agents and pharmaceutical agents for the treatment of prostate cancer
PT3122757T (en) 2014-02-28 2023-11-03 Hangzhou Dac Biotech Co Ltd Charged linkers and their uses for conjugation
US10160812B2 (en) 2014-04-11 2018-12-25 Medimmune, Llc Bispecific HER2 antibodies
US10077287B2 (en) 2014-11-10 2018-09-18 Bristol-Myers Squibb Company Tubulysin analogs and methods of making and use
US10188759B2 (en) 2015-01-07 2019-01-29 Endocyte, Inc. Conjugates for imaging
AU2016222575A1 (en) * 2015-02-25 2017-09-07 William Marsh Rice University Desacetoxytubulysin H and analogs thereof
CA2991973C (en) 2015-07-12 2021-12-07 Suzhou M-Conj Biotech Co., Ltd. Bridge linkers for conjugation of a cell-binding molecule
US9839687B2 (en) 2015-07-15 2017-12-12 Suzhou M-Conj Biotech Co., Ltd. Acetylenedicarboxyl linkers and their uses in specific conjugation of a cell-binding molecule
EP3374398B1 (en) 2015-11-10 2020-03-18 MedImmune, LLC Binding molecules specific for asct2 and uses thereof
US11793880B2 (en) 2015-12-04 2023-10-24 Seagen Inc. Conjugates of quaternized tubulysin compounds
WO2017096311A1 (en) 2015-12-04 2017-06-08 Seattle Genetics, Inc. Conjugates of quaternized tubulysin compounds
WO2017181974A1 (en) * 2016-04-20 2017-10-26 苏州苏领生物医药有限公司 Five-membered heterocyclic compound, preparation method therefor, pharmaceutical composition and use
CN107304203A (en) * 2016-04-20 2017-10-31 苏州苏领生物医药有限公司 Five-membered heterocycles and preparation method thereof, pharmaceutical composition and purposes
CN116143678A (en) 2016-11-14 2023-05-23 杭州多禧生物科技有限公司 Conjugate linker, cell-binding molecule-drug conjugate containing the same, and preparation and application thereof
MA51447A (en) 2017-08-01 2020-06-10 Medimmune Llc MONOCLONAL ANTIBODY-DRUG CONJUGATE AGAINST BCMA
WO2019108685A1 (en) 2017-11-29 2019-06-06 William March Rice University Tubulysin analogues as anticancer agents and payloads for antibody-drug conjugates and methods of treatment therewith
MA53765A1 (en) * 2018-12-21 2022-02-28 Regeneron Pharma Tubulysins and tubulysins-protein conjugates
WO2021000067A1 (en) 2019-06-29 2021-01-07 杭州多禧生物科技有限公司 Cell-binding molecule-tubulysin derivative conjugate and preparation method therefor
CA3185601A1 (en) * 2020-06-24 2021-12-30 Regeneron Pharmaceuticals, Inc. Tubulysins and protein-tubulysin conjugates
KR20230084497A (en) 2020-09-11 2023-06-13 메디뮨 리미티드 Therapeutic B7-H4 binding molecules
GB202117928D0 (en) 2021-12-11 2022-01-26 Cancer Research Tech Ltd Immunotherapy for cancer
WO2023169896A1 (en) 2022-03-09 2023-09-14 Astrazeneca Ab BINDING MOLECULES AGAINST FRα

Citations (109)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2515483A (en) 1946-08-10 1950-07-18 Merck & Co Inc Diacylated pteroic acid and process for preparing same
US2816110A (en) 1956-11-23 1957-12-10 Merck & Co Inc Methods for the production of substituted pteridines
US3387001A (en) 1964-10-19 1968-06-04 Lilly Co Eli Novel aminoacyl esters of desacetyl vincaleukoblastine
US3392173A (en) 1964-03-09 1968-07-09 Lilly Co Eli Novel acyl derivatives of desacetyl-vincaleukoblastine and processes for their preparation
US4166810A (en) 1978-04-20 1979-09-04 Eli Lilly And Company Derivatives of 4-desacetyl VLB C-3 carboxyhydrazide
US4203898A (en) 1977-08-29 1980-05-20 Eli Lilly And Company Amide derivatives of VLB, leurosidine, leurocristine and related dimeric alkaloids
US4337339A (en) 1979-04-30 1982-06-29 Baker Instruments Corp. Process for preparation of folic acid derivatives
EP0116208A1 (en) 1982-12-07 1984-08-22 Kyowa Hakko Kogyo Co., Ltd. Mitomycin analogues
JPS59175493A (en) 1983-03-25 1984-10-04 Kyowa Hakko Kogyo Co Ltd Mitomycin derivative and its preparation
JPS60255789A (en) 1984-06-01 1985-12-17 Kyowa Hakko Kogyo Co Ltd Mitomycin derivative, its preparation, and antitumor agent
US4713249A (en) 1981-11-12 1987-12-15 Schroeder Ulf Crystallized carbohydrate matrix for biologically active substances, a process of preparing said matrix, and the use thereof
WO1988001622A1 (en) 1986-08-29 1988-03-10 Kyowa Hakko Kogyo Kabusiki Kaisha Mitomycin derivatives
US4801688A (en) 1986-05-27 1989-01-31 Eli Lilly And Company Hydrazone immunoglobulin conjugates
US4866180A (en) 1984-02-24 1989-09-12 Bristol-Myers Company Amino disulfide thiol exchange products
EP0354728A2 (en) 1988-08-08 1990-02-14 Eli Lilly And Company Cytotoxic drug conjugates
WO1990012096A1 (en) 1989-04-03 1990-10-18 Purdue Research Foundation Method for enhancing transmembrane transport of exogenous molecules
WO1991007418A1 (en) 1989-11-13 1991-05-30 Xoma Corporation Chimeric mouse-human a10 antibody with specificity to a human tumor cell antigen
US5094849A (en) 1988-08-08 1992-03-10 Eli Lilly And Company Cytotoxic antibody conjugates of hydrazide derivatized vinca analogs via simple organic linkers
US5140104A (en) 1982-03-09 1992-08-18 Cytogen Corporation Amine derivatives of folic acid analogs
US5266333A (en) 1985-03-06 1993-11-30 American Cyanamid Company Water dispersible and water soluble carbohydrate polymer compositions for parenteral administration of growth hormone
US5417982A (en) 1994-02-17 1995-05-23 Modi; Pankaj Controlled release of drugs or hormones in biodegradable polymer microspheres
US5547668A (en) 1995-05-05 1996-08-20 The Board Of Trustees Of The University Of Illinois Conjugates of folate anti-effector cell antibodies
US5552545A (en) 1991-12-20 1996-09-03 Eli Lilly And Company 5-deaza-10-oxo-and 5-deaza-10-thio-5,6,7,8-tetrahydrofolic acids
WO1996036367A1 (en) 1995-05-16 1996-11-21 Purdue Research Foundation Composition and method for tumor imaging
US5627165A (en) 1990-06-13 1997-05-06 Drug Innovation & Design, Inc. Phosphorous prodrugs and therapeutic delivery systems using same
US5672486A (en) 1990-08-29 1997-09-30 Centre Hospitalier Regional De Nantes Protein polyligands joined to a stable protein core
WO1998010651A1 (en) 1996-09-12 1998-03-19 Merck & Co., Inc. Conjugates useful in the treatment of prostate cancer
WO1999020626A1 (en) 1997-10-17 1999-04-29 Purdue Research Foundation Folic acid derivatives
WO1999061055A1 (en) 1998-05-22 1999-12-02 The Board Of Trustees Of The Leland Stanford Junior University Bifunctional molecules and therapies based thereon
US5998603A (en) 1994-09-29 1999-12-07 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analogs, and oligomers thereof
US6004555A (en) 1992-03-05 1999-12-21 Board Of Regents, The University Of Texas System Methods for the specific coagulation of vasculature
US6030941A (en) 1996-05-01 2000-02-29 Avi Biopharma, Inc. Polymer composition for delivering substances in living organisms
US6056973A (en) 1996-10-11 2000-05-02 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method of preparation
US6077499A (en) 1996-05-03 2000-06-20 Immunomedics, Inc. Targeted combination immunotherapy of cancer
US6093382A (en) 1998-05-16 2000-07-25 Bracco Research Usa Inc. Metal complexes derivatized with folate for use in diagnostic and therapeutic applications
WO2000035422A3 (en) 1998-12-18 2000-10-12 Hadasit Med Res Service Method of administering a compound to multi-drug resistant cells
WO2000066091A1 (en) 1999-05-04 2000-11-09 Biotech Australia Pty Limited Amplification of folate-mediated targeting to tumor cells using polymers
CA2376175A1 (en) 1999-06-02 2000-12-14 Biotech Australia Pty Limited Vitamin directed dual targeting therapy
US6171614B1 (en) 1996-10-15 2001-01-09 Emory University Synthesis of glycophospholipid and peptide-phospholipid conjugates and uses thereof
US6171859B1 (en) 1994-03-30 2001-01-09 Mitokor Method of targeting conjugate molecules to mitochondria
US6177404B1 (en) 1996-10-15 2001-01-23 Merck & Co., Inc. Conjugates useful in the treatment of benign prostatic hyperplasia
US6184042B1 (en) 1996-05-24 2001-02-06 Boehringer Mannheim Gmbh Method for reducing hook effect in an immunoassay
US6207157B1 (en) 1996-04-23 2001-03-27 The United States Of America As Represented By The Department Of Health And Human Services Conjugate vaccine for nontypeable Haemophilus influenzae
WO2001028592A1 (en) 1999-10-15 2001-04-26 Mayo Foundation For Medical Education And Research Cobalamin conjugates useful as imaging agents and as antitumor agents
US6291684B1 (en) 1999-03-29 2001-09-18 Bristol-Myers Squibb Company Process for the preparation of aziridinyl epothilones from oxiranyl epothilones
US6335434B1 (en) 1998-06-16 2002-01-01 Isis Pharmaceuticals, Inc., Nucleosidic and non-nucleosidic folate conjugates
US6365179B1 (en) 1999-04-23 2002-04-02 Alza Corporation Conjugate having a cleavable linkage for use in a liposome
US6399625B1 (en) 2000-09-27 2002-06-04 Wyeth 1-oxorapamycins
US6399626B1 (en) 2000-10-02 2002-06-04 Wyeth Hydroxyesters of 7-desmethylrapamycin
US6399638B1 (en) 1998-04-21 2002-06-04 Bristol-Myers Squibb Company 12,13-modified epothilone derivatives
US6432973B1 (en) 2000-09-19 2002-08-13 Wyeth Water soluble rapamycin esters
US6440991B1 (en) 2000-10-02 2002-08-27 Wyeth Ethers of 7-desmethlrapamycin
WO2001074382A9 (en) 2000-03-31 2002-10-10 Purdue Research Foundation Method of treatment using ligand-immunogen conjugates
WO2002085908A1 (en) 2001-04-24 2002-10-31 Purdue Research Foundation Folate mimetics and folate-receptor binding conjugates thereof
WO2002098868A1 (en) 2001-06-01 2002-12-12 Bristol-Myers Squibb Company Epothilone derivatives
US6511986B2 (en) 2000-08-11 2003-01-28 Wyeth Method of treating estrogen receptor positive carcinoma
WO2002087424A3 (en) 2001-05-02 2003-03-20 Philip Stewart Low Treatment and diagnosis of macrophage mediated disease
US6541612B2 (en) 1993-04-23 2003-04-01 Wyeth Monoclonal antibodies obtained using rapamycin position 27 conjugates as an immunogen
US20030086900A1 (en) 2001-09-28 2003-05-08 Low Philip S. Method of treatment using ligand-immunogen conjugates
US6596757B1 (en) 2002-05-14 2003-07-22 Immunogen Inc. Cytotoxic agents comprising polyethylene glycol-containing taxanes and their therapeutic use
US20030162234A1 (en) 2002-02-07 2003-08-28 Jallad Karim N. Folate targeted enhanced tumor and folate receptor positive tissue optical imaging technology
US6617333B2 (en) 2001-08-07 2003-09-09 Wyeth Antineoplastic combinations comprising
WO2003097647A1 (en) 2002-05-15 2003-11-27 Endocyte, Inc. Vitamin-mitomycin conjugates
US6670355B2 (en) 2000-06-16 2003-12-30 Wyeth Method of treating cardiovascular disease
US6677357B2 (en) 2001-08-22 2004-01-13 Wyeth Rapamycin 29-enols
WO2004005327A1 (en) 2002-07-09 2004-01-15 Morphochem Ag Komb Chemie Novel tubulysin analogues
US6680330B2 (en) 2001-08-22 2004-01-20 Wyeth Rapamycin dialdehydes
US20040018203A1 (en) 2001-06-08 2004-01-29 Ira Pastan Pegylation of linkers improves antitumor activity and reduces toxicity of immunoconjugates
WO2004005326A3 (en) 2002-07-09 2004-02-19 Morphochem Aktiengellschaft Fu Tubulysin conjugates
US20040033195A1 (en) 2002-05-06 2004-02-19 Leamon Christopher P. Vitamin-targeted imaging agents
US6713607B2 (en) 1994-03-08 2004-03-30 Wyeth Effector proteins of Rapamycin
WO2004012735A3 (en) 2002-07-31 2004-05-27 Schering Ag New effector conjugates, process for their production and their pharmaceutical use
WO2004054622A1 (en) 2002-12-13 2004-07-01 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
WO2004046170A3 (en) 2002-11-21 2004-07-01 Biotechnolog Forschung Gmbh Tubulysins, method for producing the same and tubulysin preparations
US6821731B2 (en) 2000-11-28 2004-11-23 Wyeth Expression analysis of FKBP nucleic acids and polypeptides useful in the diagnosis of prostate cancer
US20050002942A1 (en) 2003-01-27 2005-01-06 Vlahov Iontcho R. Vitamin receptor binding drug delivery conjugates
US20050026068A1 (en) 2001-11-01 2005-02-03 Evangelos Gogolides Polycarbocyclic derivatives for modification of resist, optical and etch resistance properties
US20050107325A1 (en) 2003-04-17 2005-05-19 Muthiah Manoharan Modified iRNA agents
US6915855B2 (en) 2002-05-02 2005-07-12 Halliburton Energy Services, Inc. Wellbore junction drifting apparatus and associated method
US6958153B1 (en) 1997-11-07 2005-10-25 Wyeth Skin penetration enhancing components
US20050239739A1 (en) 2001-05-18 2005-10-27 Sirna Therapeutics, Inc. Conjugates and compositions for cellular delivery
WO2004100983A3 (en) 2003-05-06 2005-11-10 Purdue Res Foundation Inc Treatment of lupus targeting the macrophages or the folate receptor
WO2006012527A8 (en) 2004-07-23 2006-03-09 Endocyte Inc Bivalent linkers and conjugates thereof
US20060058266A1 (en) 2004-08-10 2006-03-16 Muthiah Manoharan Chemically modified oligonucleotides
US7019014B2 (en) 2003-05-12 2006-03-28 Wyeth Holdings Corporation Process for producing anticancer agent LL-D45042
WO2005074901A3 (en) 2004-01-30 2006-03-30 Schering Ag New effector conjugates, process for their production and their pharmaceutical use
US7029674B2 (en) 2001-04-02 2006-04-18 Wyeth Methods for downmodulating immune cells using an antibody to PD-1
WO2006042146A2 (en) 2004-10-07 2006-04-20 Emory University Multifunctional nanoparticles conjugates and their use
US7060797B2 (en) 2002-11-21 2006-06-13 Wyeth Composition and method for treating lupus nephritis
US7060709B2 (en) 2003-02-06 2006-06-13 Wyeth Method of treating hepatic fibrosis
US7067111B1 (en) 1999-10-25 2006-06-27 Board Of Regents, University Of Texas System Ethylenedicysteine (EC)-drug conjugates, compositions and methods for tissue specific disease imaging
US7074804B2 (en) 2003-07-16 2006-07-11 Wyeth CCI-779 Isomer C
US7105328B2 (en) 2001-04-02 2006-09-12 Dana-Farber Cancer Institute Methods for screening for compounds that modulate pd-1 signaling
WO2006105141A1 (en) 2005-03-30 2006-10-05 Purdue Research Foundation Method for cancer prognosis using cellular folate vitamin receptor quantification
US7122361B2 (en) 2002-10-10 2006-10-17 Wyeth Compositions employing a novel human kinase
WO2005112919A8 (en) 2004-05-19 2006-12-07 Medarex Inc Self-immolative linkers and drug conjugates
US7153957B2 (en) 2003-08-07 2006-12-26 Wyeth Regioselective synthesis of CCI-779
US20070009434A1 (en) 2005-07-05 2007-01-11 Low Philip S Imaging and therapeutic method using monocytes
WO2006101845A3 (en) 2005-03-16 2007-03-29 Endocyte Inc Synthesis and purification of pteroic acid and conjugates thereof
WO2007022494A3 (en) 2005-08-19 2007-09-07 Endocyte Inc Multi-drug ligand conjugates
US20070275904A1 (en) 2006-05-25 2007-11-29 Bristol-Myers Squibb Company Conjugates of aziridinyl-epothilone analogs and pharmaceutical compositions comprising same
WO2007022493A3 (en) 2005-08-19 2008-01-24 Endocyte Inc Ligand conjugates of vinca alkaloids, analogs, and derivatives
WO2008101231A2 (en) 2007-02-16 2008-08-21 Endocyte, Inc. Methods and compositions for treating and diagnosing kidney disease
WO2009002993A1 (en) 2007-06-25 2008-12-31 Endocyte, Inc. Conjugates containing hydrophilic spacer linkers
WO2009055562A1 (en) 2007-10-25 2009-04-30 Endocyte, Inc. Tubulysins and processes for preparing
WO2008112873A3 (en) 2007-03-14 2009-09-11 Endocyte, Inc. Binding ligand linked drug delivery conjugates of tubulysins
WO2011069116A1 (en) 2009-12-04 2011-06-09 Endocyte, Inc. Binding ligand linked drug delivery conjugates of tubulysins
WO2011106639A1 (en) 2010-02-25 2011-09-01 Purdue Research Foundation Psma binding ligand-linker conjugates and methods for using
WO2012019123A1 (en) 2010-08-06 2012-02-09 Endocyte, Inc. Processes for preparing tubulysins

Patent Citations (140)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2515483A (en) 1946-08-10 1950-07-18 Merck & Co Inc Diacylated pteroic acid and process for preparing same
US2816110A (en) 1956-11-23 1957-12-10 Merck & Co Inc Methods for the production of substituted pteridines
US3392173A (en) 1964-03-09 1968-07-09 Lilly Co Eli Novel acyl derivatives of desacetyl-vincaleukoblastine and processes for their preparation
US3387001A (en) 1964-10-19 1968-06-04 Lilly Co Eli Novel aminoacyl esters of desacetyl vincaleukoblastine
US4203898A (en) 1977-08-29 1980-05-20 Eli Lilly And Company Amide derivatives of VLB, leurosidine, leurocristine and related dimeric alkaloids
US4166810A (en) 1978-04-20 1979-09-04 Eli Lilly And Company Derivatives of 4-desacetyl VLB C-3 carboxyhydrazide
US4337339A (en) 1979-04-30 1982-06-29 Baker Instruments Corp. Process for preparation of folic acid derivatives
US4713249A (en) 1981-11-12 1987-12-15 Schroeder Ulf Crystallized carbohydrate matrix for biologically active substances, a process of preparing said matrix, and the use thereof
US5140104A (en) 1982-03-09 1992-08-18 Cytogen Corporation Amine derivatives of folic acid analogs
EP0116208A1 (en) 1982-12-07 1984-08-22 Kyowa Hakko Kogyo Co., Ltd. Mitomycin analogues
JPS59175493A (en) 1983-03-25 1984-10-04 Kyowa Hakko Kogyo Co Ltd Mitomycin derivative and its preparation
US4866180A (en) 1984-02-24 1989-09-12 Bristol-Myers Company Amino disulfide thiol exchange products
US4691024A (en) 1984-06-01 1987-09-01 Kyowa Hakko Kogyo Kabushiki Kaisha New mitomycin derivatives, preparation thereof and pharmaceutical compositions containing them
JPS60255789A (en) 1984-06-01 1985-12-17 Kyowa Hakko Kogyo Co Ltd Mitomycin derivative, its preparation, and antitumor agent
US5266333A (en) 1985-03-06 1993-11-30 American Cyanamid Company Water dispersible and water soluble carbohydrate polymer compositions for parenteral administration of growth hormone
US4801688A (en) 1986-05-27 1989-01-31 Eli Lilly And Company Hydrazone immunoglobulin conjugates
WO1988001622A1 (en) 1986-08-29 1988-03-10 Kyowa Hakko Kogyo Kabusiki Kaisha Mitomycin derivatives
EP0280741A1 (en) 1986-08-29 1988-09-07 Kyowa Hakko Kogyo Kabushiki Kaisha Mitomycin derivatives
EP0354728A2 (en) 1988-08-08 1990-02-14 Eli Lilly And Company Cytotoxic drug conjugates
US5094849A (en) 1988-08-08 1992-03-10 Eli Lilly And Company Cytotoxic antibody conjugates of hydrazide derivatized vinca analogs via simple organic linkers
US5006652A (en) 1988-08-08 1991-04-09 Eli Lilly And Company Intermediates for antibody-vinca drug conjugates
US5635382A (en) 1989-04-03 1997-06-03 Purdue Research Foundation Method for enhancing transmembrane transport of exogenous molecules
US5108921A (en) 1989-04-03 1992-04-28 Purdue Research Foundation Method for enhanced transmembrane transport of exogenous molecules
WO1990012096A1 (en) 1989-04-03 1990-10-18 Purdue Research Foundation Method for enhancing transmembrane transport of exogenous molecules
US5416016A (en) 1989-04-03 1995-05-16 Purdue Research Foundation Method for enhancing transmembrane transport of exogenous molecules
US5688488A (en) 1989-04-03 1997-11-18 Purdue Research Foundation Composition and method for tumor imaging
WO1991007418A1 (en) 1989-11-13 1991-05-30 Xoma Corporation Chimeric mouse-human a10 antibody with specificity to a human tumor cell antigen
US5627165A (en) 1990-06-13 1997-05-06 Drug Innovation & Design, Inc. Phosphorous prodrugs and therapeutic delivery systems using same
US5672486A (en) 1990-08-29 1997-09-30 Centre Hospitalier Regional De Nantes Protein polyligands joined to a stable protein core
US5552545A (en) 1991-12-20 1996-09-03 Eli Lilly And Company 5-deaza-10-oxo-and 5-deaza-10-thio-5,6,7,8-tetrahydrofolic acids
US6004555A (en) 1992-03-05 1999-12-21 Board Of Regents, The University Of Texas System Methods for the specific coagulation of vasculature
US6541612B2 (en) 1993-04-23 2003-04-01 Wyeth Monoclonal antibodies obtained using rapamycin position 27 conjugates as an immunogen
US5417982A (en) 1994-02-17 1995-05-23 Modi; Pankaj Controlled release of drugs or hormones in biodegradable polymer microspheres
US6713607B2 (en) 1994-03-08 2004-03-30 Wyeth Effector proteins of Rapamycin
US6171859B1 (en) 1994-03-30 2001-01-09 Mitokor Method of targeting conjugate molecules to mitochondria
US5998603A (en) 1994-09-29 1999-12-07 Isis Pharmaceuticals, Inc. 4'-desmethyl nucleoside analogs, and oligomers thereof
US5547668A (en) 1995-05-05 1996-08-20 The Board Of Trustees Of The University Of Illinois Conjugates of folate anti-effector cell antibodies
WO1996036367A1 (en) 1995-05-16 1996-11-21 Purdue Research Foundation Composition and method for tumor imaging
US6207157B1 (en) 1996-04-23 2001-03-27 The United States Of America As Represented By The Department Of Health And Human Services Conjugate vaccine for nontypeable Haemophilus influenzae
US6030941A (en) 1996-05-01 2000-02-29 Avi Biopharma, Inc. Polymer composition for delivering substances in living organisms
US6077499A (en) 1996-05-03 2000-06-20 Immunomedics, Inc. Targeted combination immunotherapy of cancer
US6184042B1 (en) 1996-05-24 2001-02-06 Boehringer Mannheim Gmbh Method for reducing hook effect in an immunoassay
WO1998010651A1 (en) 1996-09-12 1998-03-19 Merck & Co., Inc. Conjugates useful in the treatment of prostate cancer
US6056973A (en) 1996-10-11 2000-05-02 Sequus Pharmaceuticals, Inc. Therapeutic liposome composition and method of preparation
US6171614B1 (en) 1996-10-15 2001-01-09 Emory University Synthesis of glycophospholipid and peptide-phospholipid conjugates and uses thereof
US6177404B1 (en) 1996-10-15 2001-01-23 Merck & Co., Inc. Conjugates useful in the treatment of benign prostatic hyperplasia
US6291673B1 (en) 1997-10-17 2001-09-18 Purdue Research Foundation Folic acid derivatives
WO1999020626A1 (en) 1997-10-17 1999-04-29 Purdue Research Foundation Folic acid derivatives
US6958153B1 (en) 1997-11-07 2005-10-25 Wyeth Skin penetration enhancing components
US6399638B1 (en) 1998-04-21 2002-06-04 Bristol-Myers Squibb Company 12,13-modified epothilone derivatives
US6093382A (en) 1998-05-16 2000-07-25 Bracco Research Usa Inc. Metal complexes derivatized with folate for use in diagnostic and therapeutic applications
WO1999061055A1 (en) 1998-05-22 1999-12-02 The Board Of Trustees Of The Leland Stanford Junior University Bifunctional molecules and therapies based thereon
US6335434B1 (en) 1998-06-16 2002-01-01 Isis Pharmaceuticals, Inc., Nucleosidic and non-nucleosidic folate conjugates
WO2000035422A3 (en) 1998-12-18 2000-10-12 Hadasit Med Res Service Method of administering a compound to multi-drug resistant cells
US6291684B1 (en) 1999-03-29 2001-09-18 Bristol-Myers Squibb Company Process for the preparation of aziridinyl epothilones from oxiranyl epothilones
US6365179B1 (en) 1999-04-23 2002-04-02 Alza Corporation Conjugate having a cleavable linkage for use in a liposome
WO2000066091A1 (en) 1999-05-04 2000-11-09 Biotech Australia Pty Limited Amplification of folate-mediated targeting to tumor cells using polymers
CA2372841A1 (en) 1999-05-04 2000-11-09 Gregory John Russell-Jones Amplification of folate-mediated targeting to tumor cells using polymers
CA2376175A1 (en) 1999-06-02 2000-12-14 Biotech Australia Pty Limited Vitamin directed dual targeting therapy
WO2000074721A1 (en) 1999-06-02 2000-12-14 Biotech Australia Pty Limited Vitamin directed dual targeting therapy
WO2001028592A1 (en) 1999-10-15 2001-04-26 Mayo Foundation For Medical Education And Research Cobalamin conjugates useful as imaging agents and as antitumor agents
US20050004010A1 (en) 1999-10-15 2005-01-06 Mayo Foundation For Medical Education Cobalamin conjugates useful as imaging agents and as antitumor agents
US7067111B1 (en) 1999-10-25 2006-06-27 Board Of Regents, University Of Texas System Ethylenedicysteine (EC)-drug conjugates, compositions and methods for tissue specific disease imaging
WO2001074382A9 (en) 2000-03-31 2002-10-10 Purdue Research Foundation Method of treatment using ligand-immunogen conjugates
US7033594B2 (en) 2000-03-31 2006-04-25 Purdue Research Foundation Method of treatment using ligand-immunogen conjugates
US6670355B2 (en) 2000-06-16 2003-12-30 Wyeth Method of treating cardiovascular disease
US6511986B2 (en) 2000-08-11 2003-01-28 Wyeth Method of treating estrogen receptor positive carcinoma
US6432973B1 (en) 2000-09-19 2002-08-13 Wyeth Water soluble rapamycin esters
US6399625B1 (en) 2000-09-27 2002-06-04 Wyeth 1-oxorapamycins
US6440991B1 (en) 2000-10-02 2002-08-27 Wyeth Ethers of 7-desmethlrapamycin
US6399626B1 (en) 2000-10-02 2002-06-04 Wyeth Hydroxyesters of 7-desmethylrapamycin
US6821731B2 (en) 2000-11-28 2004-11-23 Wyeth Expression analysis of FKBP nucleic acids and polypeptides useful in the diagnosis of prostate cancer
US7029674B2 (en) 2001-04-02 2006-04-18 Wyeth Methods for downmodulating immune cells using an antibody to PD-1
US7105328B2 (en) 2001-04-02 2006-09-12 Dana-Farber Cancer Institute Methods for screening for compounds that modulate pd-1 signaling
US20040242582A1 (en) 2001-04-24 2004-12-02 Green Mark A Folate mimetics and folate-receptor binding conjugates thereof
US20050227985A9 (en) 2001-04-24 2005-10-13 Green Mark A Folate mimetics and folate-receptor binding conjugates thereof
WO2002085908A1 (en) 2001-04-24 2002-10-31 Purdue Research Foundation Folate mimetics and folate-receptor binding conjugates thereof
WO2002087424A3 (en) 2001-05-02 2003-03-20 Philip Stewart Low Treatment and diagnosis of macrophage mediated disease
US20050239739A1 (en) 2001-05-18 2005-10-27 Sirna Therapeutics, Inc. Conjugates and compositions for cellular delivery
WO2002098868A1 (en) 2001-06-01 2002-12-12 Bristol-Myers Squibb Company Epothilone derivatives
US6800653B2 (en) 2001-06-01 2004-10-05 Bristol-Myers Squibb Compnay Epothilone derivatives
US20040018203A1 (en) 2001-06-08 2004-01-29 Ira Pastan Pegylation of linkers improves antitumor activity and reduces toxicity of immunoconjugates
US6617333B2 (en) 2001-08-07 2003-09-09 Wyeth Antineoplastic combinations comprising
US6680330B2 (en) 2001-08-22 2004-01-20 Wyeth Rapamycin dialdehydes
US6677357B2 (en) 2001-08-22 2004-01-13 Wyeth Rapamycin 29-enols
US20030086900A1 (en) 2001-09-28 2003-05-08 Low Philip S. Method of treatment using ligand-immunogen conjugates
US20050026068A1 (en) 2001-11-01 2005-02-03 Evangelos Gogolides Polycarbocyclic derivatives for modification of resist, optical and etch resistance properties
US20030162234A1 (en) 2002-02-07 2003-08-28 Jallad Karim N. Folate targeted enhanced tumor and folate receptor positive tissue optical imaging technology
US6915855B2 (en) 2002-05-02 2005-07-12 Halliburton Energy Services, Inc. Wellbore junction drifting apparatus and associated method
US7128893B2 (en) 2002-05-06 2006-10-31 Endocyte, Inc. Vitamin-targeted imaging agents
US20040033195A1 (en) 2002-05-06 2004-02-19 Leamon Christopher P. Vitamin-targeted imaging agents
US6596757B1 (en) 2002-05-14 2003-07-22 Immunogen Inc. Cytotoxic agents comprising polyethylene glycol-containing taxanes and their therapeutic use
WO2003097647A1 (en) 2002-05-15 2003-11-27 Endocyte, Inc. Vitamin-mitomycin conjugates
US20050165227A1 (en) 2002-05-15 2005-07-28 Vlahov Iontcho R. Vitamin-mitomycin conjugates
US7816377B2 (en) 2002-07-09 2010-10-19 R&D-Biopharmaceuticals Gmbh Tubulysin analogues
WO2004005327A1 (en) 2002-07-09 2004-01-15 Morphochem Ag Komb Chemie Novel tubulysin analogues
WO2004005326A3 (en) 2002-07-09 2004-02-19 Morphochem Aktiengellschaft Fu Tubulysin conjugates
US20050239713A1 (en) 2002-07-09 2005-10-27 R&D-Biopharmaceuticals Am Novel tubulysin analogues
US7776814B2 (en) 2002-07-09 2010-08-17 R&D-Biopharmaceuticals Gmbh Tubulysin conjugates
WO2004012735A3 (en) 2002-07-31 2004-05-27 Schering Ag New effector conjugates, process for their production and their pharmaceutical use
US7122361B2 (en) 2002-10-10 2006-10-17 Wyeth Compositions employing a novel human kinase
US20060128754A1 (en) * 2002-11-21 2006-06-15 Gerhard Hoefle Tubulysins, method for producing the same and tubulysin preparations
US7060797B2 (en) 2002-11-21 2006-06-13 Wyeth Composition and method for treating lupus nephritis
WO2004046170A3 (en) 2002-11-21 2004-07-01 Biotechnolog Forschung Gmbh Tubulysins, method for producing the same and tubulysin preparations
US7754885B2 (en) 2002-11-21 2010-07-13 Helmholtz-Zentrum Fuer Infektionsforschung Gmbh Tubulysins, method for producing the same and tubulysin preparations
WO2004054622A1 (en) 2002-12-13 2004-07-01 Immunomedics, Inc. Immunoconjugates with an intracellularly-cleavable linkage
WO2004069159A3 (en) 2003-01-27 2005-06-16 Endocyte Inc Vitamin receptor binding drug delivery conjugates
US20050002942A1 (en) 2003-01-27 2005-01-06 Vlahov Iontcho R. Vitamin receptor binding drug delivery conjugates
US20100004276A1 (en) 2003-01-27 2010-01-07 Vlahov Iontcho R Vitamin receptor binding drug delivery conjugates
US7601332B2 (en) 2003-01-27 2009-10-13 Endocyte, Inc. Vitamin receptor binding drug delivery conjugates
US7060709B2 (en) 2003-02-06 2006-06-13 Wyeth Method of treating hepatic fibrosis
US20050107325A1 (en) 2003-04-17 2005-05-19 Muthiah Manoharan Modified iRNA agents
WO2004100983A3 (en) 2003-05-06 2005-11-10 Purdue Res Foundation Inc Treatment of lupus targeting the macrophages or the folate receptor
US7019014B2 (en) 2003-05-12 2006-03-28 Wyeth Holdings Corporation Process for producing anticancer agent LL-D45042
US7074804B2 (en) 2003-07-16 2006-07-11 Wyeth CCI-779 Isomer C
US7153957B2 (en) 2003-08-07 2006-12-26 Wyeth Regioselective synthesis of CCI-779
WO2005074901A3 (en) 2004-01-30 2006-03-30 Schering Ag New effector conjugates, process for their production and their pharmaceutical use
WO2005112919A8 (en) 2004-05-19 2006-12-07 Medarex Inc Self-immolative linkers and drug conjugates
WO2006012527A8 (en) 2004-07-23 2006-03-09 Endocyte Inc Bivalent linkers and conjugates thereof
US20090203889A1 (en) 2004-07-23 2009-08-13 Endocyte, Inc. Bivalent linkers and conjugates thereof
US20060058266A1 (en) 2004-08-10 2006-03-16 Muthiah Manoharan Chemically modified oligonucleotides
WO2006042146A2 (en) 2004-10-07 2006-04-20 Emory University Multifunctional nanoparticles conjugates and their use
WO2006101845A3 (en) 2005-03-16 2007-03-29 Endocyte Inc Synthesis and purification of pteroic acid and conjugates thereof
US20080207625A1 (en) 2005-03-16 2008-08-28 Endocyte, Inc. Synthesis and Purification of Pteroic Acid and Conjugates Thereof
WO2006105141A1 (en) 2005-03-30 2006-10-05 Purdue Research Foundation Method for cancer prognosis using cellular folate vitamin receptor quantification
US20070009434A1 (en) 2005-07-05 2007-01-11 Low Philip S Imaging and therapeutic method using monocytes
WO2007022493A3 (en) 2005-08-19 2008-01-24 Endocyte Inc Ligand conjugates of vinca alkaloids, analogs, and derivatives
US20080280937A1 (en) 2005-08-19 2008-11-13 Christopher Paul Leamon Ligand Conjugates of Vinca Alkaloids, Analogs, and Derivatives
US20080248052A1 (en) 2005-08-19 2008-10-09 Iontcho Radoslavov Vlahov Multi-Drug Ligand Conjugates
WO2007022494A3 (en) 2005-08-19 2007-09-07 Endocyte Inc Multi-drug ligand conjugates
US20070275904A1 (en) 2006-05-25 2007-11-29 Bristol-Myers Squibb Company Conjugates of aziridinyl-epothilone analogs and pharmaceutical compositions comprising same
WO2008101231A2 (en) 2007-02-16 2008-08-21 Endocyte, Inc. Methods and compositions for treating and diagnosing kidney disease
US20100104626A1 (en) 2007-02-16 2010-04-29 Endocyte, Inc. Methods and compositions for treating and diagnosing kidney disease
US20100048490A1 (en) 2007-03-14 2010-02-25 Iontcho Radoslavov Vlahov Binding ligand linked drug delivery conjugates of tubulysins
WO2008112873A3 (en) 2007-03-14 2009-09-11 Endocyte, Inc. Binding ligand linked drug delivery conjugates of tubulysins
WO2009002993A1 (en) 2007-06-25 2008-12-31 Endocyte, Inc. Conjugates containing hydrophilic spacer linkers
WO2009055562A1 (en) 2007-10-25 2009-04-30 Endocyte, Inc. Tubulysins and processes for preparing
WO2011069116A1 (en) 2009-12-04 2011-06-09 Endocyte, Inc. Binding ligand linked drug delivery conjugates of tubulysins
WO2011106639A1 (en) 2010-02-25 2011-09-01 Purdue Research Foundation Psma binding ligand-linker conjugates and methods for using
WO2012019123A1 (en) 2010-08-06 2012-02-09 Endocyte, Inc. Processes for preparing tubulysins

Non-Patent Citations (307)

* Cited by examiner, † Cited by third party
Title
Achilefu et al. "A New Method for the Synthesis of Tri-tert-butyl Diethylenetriaminepentaacetic Acid Its Derivatives" J. Org. Chem. 2000;65:1562-1565.
Agoston E.S. et al., "Vitamin D Analogs as Anti-Carcinogenic Agents," Anti-Cancer Agents in Medicinal Chemistry, 2006; 6(1): 53-71.
Anderson et al., "Potocytosis: Sequestration and transport of small molecules by caveolae," Science, 1992; 255: 410-411.
Angier, R. B., et al., "Pteroic Acid Analogs Containing Arsenic," J. American Chem. Soc., vol. 76, 1954, pp. 902-904.
Antony A.C. et al., "Studies of the Role of a Particulate Folate-binding Protein in the Uptake of 5-Methyltetrahydrofolate by Cultured Human KB Cells," J. Biological Chem., 1985; 260(28):14911-7.
Antony A.C., "Folate receptors," Annu Rev Nutr, 1996; 16: 501-21.
Antony A.C., "The biological chemistry of folate receptors," Blood, 1992; 79(11):2807-2820.
Antony, "Folate receptors: reflections on a personal odysssey and a perspective on unfolding truth," Advanced Drug Delivery Reviews, 2004; vol. 56: 1059-66.
Archer M.C. et al., "Separation of Folic Acid Derivatives and Pterins by High-Performance Liquid Chromatography," Methods In Enzymology, 1980; 66. pp: 452-459.
Arya et al., "Design and Synthesis of Analogs of Vitamin E: Antiproliferative Activity Against Human Breast Adenocarcinoma Cells," Bioorganic & Medicinal Chemistry Letters, 1998; vol. 8, pp. 2433-2438.
Ayers W.A., "Effect of Vitamin B12 and Analogs on the Respiration of a Marine Bacterium," Archives of Biochemistry and Biophysics, 1962, vol. 96, pp. 210-215.
Barnett C.J. et al., "Structure-Activity Relationships of Dimeric Catharanthus Alkaloids. 1. Deacetylvinblastine Amide (Vindesine) Sulfate," J. Med. Chem. 21: 88-96 (1978).
Bartels R. et al., "Determination of pteroic acid by high-performance thin-layer chromatography: Contribution to the investigation of 7,8-dihydropteroate synthase," Journal of Chromatography A, 1994; vol. 659(1): 185-189 (abstract only).
Bavetsias, V. et al., "Design and synthesis of Cyclopenta[g]quinazoline-based antifolates as inhibitors of thymidylate synthase and potential antitumor agents," J Med Chem, 2000; 43(10): 1910-1926.
Bavetsias, V., et al., "The design and synthesis of water-soluble analogues of CB30865, a quinazolin-4-one-based antitumor agent," J Med Chem, 2002; 45(17): 3692-3702.
Beevers, Christopher S., et al., "Curcumin Inhibits the Mammalian Target of Rapamycin-Mediated Signaling Pathways in Cancer Cells", 2006; Int. Journal Cancer; Vo. 119; pp. 757-764.
Birinberg E. M. et al., "Synthesis and antimetabolic activity of pyrimidine analogs of folic and pteroic acids," Pharmaceutical Chemistry Journal, 1969; 3(6): pp. 331-333.
Bock et al., "Sulfonamide structure-activity relationships in a cell-free system. 2. Proof for the formation of a sulfonamide-containing folate analog," Journal of Medical Chemistry, 17: 23-28 (1974).
Boger, D.L. et al., "An improved synthesis of 1,2,9,9a-tetrahydrocyclopropa[c]benz[e]indo1-4-one (CBI): a simplified analog of the CC-1065 alkylation subunit," J. Org. Chem., 1992; 57: 2873-2876.
Boothe, J. H., et al., "Pteroic Acid Derivatives. II. Pteroyl-γ-glutamylglutamic Acid and Pteroyl-γ-glutamylyγglutamylglutamic Acid," J. American Chem. Soc., vol. 70, 1948, pp. 1099-1102.
Brown, Nicole E., et al., "Delayed Cystogenesis and Increased Ciliogenesis Associated with th Re-Expression of Polaris in Tg737 Mutant Mice", 2003, Kidney International, vol. 63, pp. 1220-1229.
Bukanov Nikolay, O. et al., "Long-Lasting Arrest of Murine Polycystic Kidney Disease With CDK Inhibitor Roscovitine", Dec. 14, 2006; Nature; vol. 444; pp. 949-952.
Campbell et al., "Folate-binding protein is a marker for ovarian cancer," Cancer Res., 1991; 51: 5329-5338.
Carl et al. "A novel connector linkage applicable in prodrug design" J. Med. Chem. 1981;24(5):479-480.
Cho et al., "Single-chain Fv/folate conjugates mediate efficient lysis of folate-receptor-positive tumor cells," Bioconjug. Chem. 8(3): 338-346 (1997).
Christensen et al., "Membrane receptors for endocytosis in the renal proximal tubule," Int. Rev. Cytol., 1998; 180: 237-284.
Churlaud, Carine, et al., "Novel 4-(Trimethylsily1)Aminoalkanes and 4-(Trimethylsily1)Aminoalk-2-enes, via a 1,5-Hydride Shift, in the Reaction of alpha-Unsaturated Silanes With Aminomethylbenzotriazoles", 1998, J. Organomet. Chem., pp. 4270-4274.
Churlaud, Carine, et al., "Novel 4-(Trimethylsily1)Aminoalkanes and 4-(Trimethylsily1)Aminoalk-2-enes, via a 1,5-Hydride Shift, in the Reaction of α-Unsaturated Silanes With Aminomethylbenzotriazoles", 1998, J. Organomet. Chem., pp. 4270-4274.
Churlaud, Carine, et al., "Novel 4-(Trimethylsilyl)Aminoalkanes and 4-(Trimethylsily1)Aminoalk-2-enes, via a 1,5-Hydride Shift, in the Reaction of α-Unsaturated Silanes With Aminomethylbenzotriazoles", 1998, J. Organomet. Chem., pp. 4270-4274.
Citro G. et al., "Inhibition of leukemia cell proliferation by folic acid-polylysine-mediated introduction of c-myb antisense oligodeoxynucelotides into HL-60 cells," Br. J. Cancer, 1994; 69: 463-467.
Coney et al. "Cloning of a tumor-associated antigen!: MOv18 and MOv19 antibodies recognize a folate-binding protein" Cancer Res. 1991;51(22):6125-32.
Cope A.C. et al., "Thermal Rearrangement of Allyl-type Sulfoxides, Sulfones and Sulfinates," J. Am. Chem. Soc., 1950; 72; 59-67.
Cosulich D.B. et al., "Analogs of Pteroylglutamic Acid. I. N10-Alkylpteroic Acid and Derivatives," JACS, 1948, 70 (5), pp. 1922-1926.
Crapatureanu et al. "Molecular necklaces. Cross-linking hemoglobin with reagents containing covalently attached ligands" Bioconjugate Chemistry, 1999;10(6):1058-67.
Darnell, Ed. Molecular Cell Biology W. H. Freeman, San Francisco 1990;326-333.
DeNardo, Gerald. "When is Too Much Too Much and Yet Not Enough? Alas, a Plethora of Opportunities but Where's the Beef?" J. of Nuclear Medicine 2000; 41(3):470-3.
DeVita, Jr., Vincent et al (eds); Biologic Therapy of Cancer; 2nd ed., J.B. Lippincott Company; 1995.
Domling A. et al., "Myxobacterial Epothilones and Tubulysins as Promising Anticancer Agents," Molecular Diversity, 2005; 9: 141-147.
Domling, A., et al., "Myxobacterial epothilones and tubulysins as promising anticancer agents," Mol. Diversity, 9:141-147 (2005).
Dorwald, F. Z., "Side Reactions in Organic Synthesis: A Guide to Successful Synthesis Design," Wiley-VCH, Weinheim, 2005, p. ix of preface.
Douglas J.T. et al., "Targeted Gene Delivery by Tropism-Modified Adenoviral Vectors," Nat. Biotechnol., 1996, vol. 14, pp. 1574-1578.
Eichman, J.D. et al., "The Use of PAMAM Dendrimers in The Efficient Transfer of Genetic Material Into Cells", Jul. 2000, PSTT, vol. 3, No. 7, pp. 232-245.
Elnakat et al., "Distribution, functionality and gene regulation of folate receptor isoforms: implications in targeted therapy," Advanced Drug Delivery Reviews, 2004; vol. 56:1067-84.
European Search Report prepared for corresponding European Application Serial No. 08841521.1, mailed Jul. 18, 2011.
Foong, L.Y. et al., "Development of a Novel Thiol Reagent for Probing Ion Channel Structure: Studies in a Model System," Biochemistry, 1997, vol. 36, pp. 1343-1348.
Forgac. "Structure and function of vacuolar class of ATP-driven pumps" Physiological Rev. 1989; 69(3): 765-795.
Frankel AE., "Immunotoxin therapy of cancer," Oncology, 1993; 7(5): 69-78.
Gabizon et al., "Tumor cell targeting of liposome-entrapped drugs with phospholipid-anchored folic acid-PEG Conjugates," Advanced Drug Delivery Reviews, 2004; vol. 56: 1177-92.
Gangjee et al., "The effect of 5-alkyl modification on the biological activity of pyrrolo[2,3-d]pyrimidine containing classical and nonclassical antifolates as inhibitors of dihydrofolate reductase and as antitumor and/or antiopportunistic infection agents," J Med Chem., 2008; 51(15):4589-4600.
Garin-Chesa et al., "Trophoblast and ovarian cancer antigen LK26. Sensitivity and specificity in immunopathology and molecular identification as a folate-binding protein," Am. J. Pathol. 142(2): 557-562 (1993).
Garrett et al. "Synthesis and characterisation of polyamine-poly(ethylene glycol) constructs for DNA binding and gene delivery" Bioorganic & Medicinal Chemistry, 2000; 8(7):1779-1797.
GE Healthcare, Instructions 71-7104-00 AD.
Gibbs, DD et al., "BGC 945, a novel tumor-selective thymidylate synthase inhibitor targeted to alpha-folate receptor-overexpressing tumors," Cancer Res, 2005; 65(24): 11721-11728.
Gottschalk S. et al., "Folate receptor mediated DNA delivery into tumor cells: potosomal disruption results in enhanced gene expression," Gene Therapy, 1994; 1(3): 185-191.
Greene T.E. et al., "Protective Groups in Organic Synthesis," 2d edition, John Wiley & Sons, Inc. New York (1991).
Griesser UJ, "The Importance of Solvents," in Polymorphism in the Pharmaceutical Industry, Hilfiker ed., 2006; p. 211-230.
Hanck A.B. et al., "Dexpanthenol (Ro 01-4709) in the treatment of constipation," Acta Vitaminol Enzymol, 1982; vol. 4 (1-2), pp. 87-97 (abstract only).
Harvison, P.J. et al., "Synthesis and Biological Activity of Novel Folic Acid Analogues: Pteroyl-S-alkylhomocysteine Sulfoximines," Journal of Medicinal Chemistry, 1992, vol. 35, pp. 1227-1233.
Hay, Nissim, et al., "Upstream and Downstream of mTOR", 2004, Genes & Development, vol. 18, No. 16, pp. 1926-1945.
Henderson et al. "Mediated uptake of folate by a high-affinity binding protein in sublines of L1210 cells adapted to nanomolar concentrations of folate" J. Membrane Biol., 1988;101:247-258.
Henderson, E.A. et al., Targeting the alpha-folate receptor with cyclopenta[g]quinazoline-based inhibitors of thymidylate synthase, Bioorg Med Chem, 2006; 14(14): 5020-5042.
Ho R. I. et al., "A simple radioassay for dihydrofolate synthetase activity in Escherichia coli and its application to an inhibition study of new pteroate analogs," Anal. Biochem., 1976, 73(2), pp. 493-500.
Hofland et al., "Folate-targeted gene transfer in vivo," Mol Ther 5(6): 739-744 (2002).
Hofle, G. et al., "Semisynthesis and Degradation of the Tubulin Inhibitors Epothilone and Tubulysin", 2003, Pure Appl. Chem., vol. 75, Nos. 2-3, pp. 167-178.
Holladay et al., "Riboflavin-mediated delivery of a macromolecule into cultured human cells," Biochim Biophys Acta, 1426(1): 195-204 (1999).
Holm, J. et al., "Folate receptors in malignant and benign tissues of human female genital tract," BioSci. Rep., 17(4): 415-427 (1997).
Holm, J. et al., "High-affinity folate binding in human choroid plexus. Characterization of radioligand binding, immunoreactivity, molecular heterogeneity and hydrophobic domain of the binding protein," Biochem J., 280(1): 267-271 (1991).
Hosomi A. et al., "Affinity for a-tocopherol transfer protein as a determinant of the biological activities of vitamin E analogs," Federation of European Biochemical Societies Letters, 1997, vol. 409, pp. 105-108.
Houlihan, C. M. et al., "Preparation and Purification of Pteroic Acid from Folic Acid," Analytical Biochemistry, 1972, vol. 46, pp. 1-6.
Huang et al. "Design, syntheses and sequence selective DNA cleavage of functional models of bleomycin-II. 1,2 -trans-di substituted cyclopropane units as novel linkers" Bioorganic & Medicinal Chemistry, 1995;3(6):647-57.
Hynes et al., "Quinazolines as inhibitors of dihydrofolate reductase. 4. Classical analogues of folic and isofolic acids", Journal of Medical Chemistry, 1977; 20: 588-591.
International Search Report Written Opinion for PCT/US2008/080948 completed Dec. 4, 2008.
Jackman, A. L. et al., "Antifolates targeted specifically to the folate receptor," Adv Drug Deliv Rev, 2004; 56(8): 1111-1125.
Jansen et al. "The Reduced Folate/Methotrexate Carrier and a Membrane-Associated Folate Binding Protein as Transport Routes for Novel Antifolates: Structure-Activity Relationship" Chemistry and Biology of Pteridines and Folates New York, 1992;767-770.
Jansen et al., "Identification of a Membrane-Associated Folate-Binding Protein in Human Leukemic CCRF-CEM Cells with Transport-Related Methotrexate Resistance" In Cancer Res., 1989, 49, 2455-2459.
Jansen, "Receptor- and Carrier-Mediated Transport Systems for Folates and Antifolates," Antifolate Drugs in Cancer Therapy, Jackman, Ed., Humana Press Inc, Totowa NJ (1999): 293-321.
Jones T.R. et al., "A potent antitumour quinazoline inhibitor of thymidylate synthetase: synthesis, biological properties and therapeutic results in mice," Eur J Cancer, 1981; 17(1):11-9.
Jones T.R. et al., "Quinazoline antifolates inhibiting thymidylate synthase: variation of the amino acid," J Med Chem, 1986; 29(6):1114-8.
Jung K.H. et al., "Intramolecular o-glycoside bond formation," Chem. Rev., 2000, 100, 4423-42.
Kagechika H et al., "Synthetic Retinoids: Recent Developments Concerning Structure and Clinical Utility," Journal of Medicinal Chemistry, 2005; vol. 48, No. 19, pp. 5875-5883.
Kamao M. et al., "Determination of Plasma Vitamin K by High Performance Liquid Chromatography with Fluorescence Detection Using Vitamin K Analogs as Internal Standards," Journal of Chromatography B, 2005; vol. 816, pp. 41-48.
Kamen et al., "A review of folate receptor alpha cycling and 5-methyltetrahydrofolate accumulation with an emphasis on cell models in vitro," Advanced Drug Delivery Reviews, 2004; vol. 56:1085-97.
Kamen et al., "Delivery of folates to the cytoplasm fo MA104 cells is mediated by a surface receptor that recycles," J. Biol. Chem., 263: 13602-13609 (1988).
Kamen et al., "The folate receptor works in tandem with a probenecid-sensitive carrier in MA104 cells in vitro," J. Clin. Invest., 87(4): 1442-1449 (1991).
Kamen, B. A. et al., "Receptor-mediated folate accumulation is regulated by the cellular folate content," Proc. Natl. Acad. Sci. USA, 83: 5983-5987 (1986).
Kandiko C.T. et al., "Inhibition of Rat Brain Pyruvate Dehydrogenase by Thiamine Analogs," Biochemical Pharmacology, 1988; vol. 37, No. 22, pp. 4375-4380.
Kane et al., "The influence of extracellular folate concentration on methotrexate uptake by human KB cells. Partial characterization of a membrane-associated methotrexate binding protein," J. Biol. Chem., 261: 44-49 (1986).
Kaur, G., et al., "Biological evaluation of tubulysin A: a potential anticancer and antiangiogenic natural product," Biochem. J. 396, 235-242 (2006).
Ke CY et al., "Folate-Receptor-Targeting of In-111 Using Pteroic Acid Conjugates of Benzyl-DTPA and Benzyl-DOTA," J. Nucl. Med., 2004; 45(5):457P.
Ke et al. "Targeting the Tumor-Associated Folate Receptor with a 111-IN-DTPA Conjugate of Pteroic Acid" Abstract No. 427. 48'h Annual Meeting of the Society of Nuclear Medicine Toronto, Canada, Jun. 26, 2001, available May 4, 2001; 1 pg.
Ke et al., "Folate-receptor-targeted radionuclide imaging agents," Advanced Drug Delivery Reviews, 2004; vol. 56: 1143-60.
Kemp et al. "New Protective Groups for Peptide Synthesis-I The Bic Group Base and Solvent Lability of the 5-B enzi soxazolymethyl eneoxycarbonyl amino function" Tet. Lett. 1975;52:4625-4628.
Kennedy, Michael D., et al., "Evaluation of Folate Conjugate Uptake and Transport by the Choroid Plexus of Mice", (May 2003), vol. 20, No. 5, pp. 714-719.
Kim et al., "Synthesis and biological activity of 10-thia-10-deaza analogs of folic acid, pteroic acid, and related compounds", Journal of Medical Chemistry, 18: 776-780 (1975).
Kranz et al., "Conjugates of folate and anti-T-cell-receptor antibodies specifically target folate-receptor-positive tumor cells for lysis," Proc. Natl. Acad. Sci. USA, 1995; 92(20), pp. 9057-9061.
Kumar H.P. et al., "Folate transport in Lactobacillus salivarius. Characterization of the transport mechanism and purification and properties of the binding component," J. Biol. Chem.. 1987; 262(15):7171-7179.
Kutsky RJ. Handbook of Vitamins, Minerals, and Hormones, 2″ Edition. New York: Van Nostrand Reinhold: 1981;263-277.
Ladino et al., "Folate-maytansinoids: target-selective drugs of low molecular weight," Int. J. Cancer, 73(6): 859 864 (1997).
Lambooy J. P., "Riboflavin Analogs Utilized for Metabolism by a Lactobacillus casei Mutant," Int. J. Biochem., vol. 16, No. 2, 1984, pp. 231-234.
Landuer W. et al., "The Interaction in Teratogenic Activity of the Two Niacin Analogs 3-acetylpyridine and 6-aminonicotinamide," J Exp Zool, 151(3):253-258 (1962).
Langone, J.J., et al., "Radioimmunoassays for the Vinca Alkaloids, Vinblastine and Vincristine", 1979, Analytical Biochemistry, No. 95, pp. 214-221.
Larock R.C., "Comprehensive Organic Transformations, a guide to functional group preparations," VCH Publishers, Inc. New York (1989).
Leamon Christopher P., "Aspects of Folate-Mediated Drug Delivery . . . Beyond Purdue" PowerPoint Presentation presented at Purdue University on May 4, 1999, (22 pages).
Leamon CP et al, "Cytotoxicity of folate-Pseudomonas exotoxin conjugates toward tumor cells. Contribution of translocation domain," J. Biol. Chem. 268(33): 24847-24854 (1993).
Leamon CP et al., "Comparative Preclinical Activity of the Folate-targeted Vinca Alkaloid Conjugates EC140 and EC145," Int J Cancer, 2007; 121(7):1585-92.
Leamon CP et al., "Cytotoxicity of momordin-folate conjugates in cultured human cells," J. Biol. Chem., 1992; 267(35): 24966-24971.
Leamon CP et al., "Delivery of macromolecules into living cells: a method that exploits folate receptor endocytosis," Proc. Natl. Acad. Sci. USA 88(13): 5572-5573 (1991).
Leamon CP et al., "Folate-mediated targeting: from diagnostics to drug and gene delivery," Drug Discovery Today 6: 36-43 (2001).
Leamon CP et al., "Folate-targeted chemotherapy," Adv Drug Deliv Rev, 2004;56(8): 1127-41.
Leamon CP et al., "Membrane folate-binding proteins are responsible for folate-protein conjugate endocytosis into cultured cells," Biochem. J. 291: 855-860 (1993).
Leamon CP et al., "Selective targeting of malignant cells with cytotoxin-folate conjugates," J. Drug Target. 2(2): 101-112 (1994).
Leamon CP et al., "Synthesis and biological evaluation of EC140: A novel folate-targeted vinca alkaloid conjugate," Bioconjug Chem, 2006;17(5):1226-32.
Leamon CP et al., "Synthesis and Biological Evaluation of EC20: A New Folate-Derived, (99m)Tc-Based Radiopharmaceutical," Bioconjug. Chem. 13(6): 1200-1210 (2002).
Leamon CP et al., "Synthesis and biological evaluation of EC72: a new folate-targeted chemotherapeutic," Bioconjug Chem., 2005;16(4):803-11.
Leamon et al., "Folate-mediated drug delivery: effect of alternative conjugation chemistry," J. Drug Target 7(3): 157-169 (1999).
Leamon, Christopher P., et al., "Folate-Liposome-Mediated Antisense Oligodeoxynucleotide Targeting to Cancer Cells: Evaluation in Vitro and in Vivo", 2003, Bioconjugate Chemistry, vol. 14, No. 4, pp. 738-747.
Lee et al, "Measurement of Endosome pH Following Folate Receptor-Mediated Endocytosis," Biochim. Biophys. Acta 1312(3): 237-242 (1996).
Lee et al. "Prolonged circulating lives of single-chain Fv proteins conjugated with polyethylene glycol: a comparison of conjugation chemistries and compounds" Bioconjugate Chemistry, 1999;10(6):973-81.
Lee et al., "BMS-247550: a novel epothilone analog with a mode of action similar to paclitaxel but possessing superior antitumor efficacy" Clin Cancer Res 7:1429-1437 (2001).
Lee et al., "Synthesis and evaluation of taxol-folic acid conjugates as targeted antineoplastics," Bioorg Med Chem. 10(7): 2397-2414, (2002).
Lee JW et al., "Reduction of azides to primary amines in substrates bearing labile ester functionality. Synthesis of a PEG-solubilized, "Y"-shaped iminodiacetic acid reagent for preparation of folate-tethered drugs," Organic Letters, 1999; 1(2):179-181.
Lee W.W. et al., "Folic acid antagonists. Methotrexate analogs containing spurious amino acids. Dichlorohomofolic acid," Journal of Medical Chemistry, 17: 326-330 (1974).
Lee, Francis Y., et al., "BMS-247550: A Novel Epothilone Analog With a Mode of Action Similar to Paclitaxel but Possessing Superior Antitumor Efficacy" 2001, Clin Cancer Res, vol. 7, pp. 1429-1437.
Lee, R. J. and Huang, L., "Folate-Targeted, Anionic Liposome-Entrapped Polylysine-Condensed Dna for Tumor Cell-Specific Gene Transfer," J. Biol. Chem. 271(14): 8481-8487 (1996).
Lee, R. J. and Low, P. S, "Delivery of liposomes into cultured KB cells via folate receptor-mediated endocytosis," J. Biol. Chem. 269(5): 3198-3204 (1994).
Lee, R. J. and Low, P. S., "Folate-mediated tumor cell targeting of liposome-entrapped doxorubicin in vitro," Biochim. Biophys. Acta 1233: 134-144 (1995).
Lemon, Julia, et al., "Conversion of Pterolyglutamic Acid to Pteroic Acid by Bacterial Degradation," Archives of Biochemistry, 1948; vol. 19, pp. 311-316.
Levy, Carl C., et al. "The Enzymatic Hydrolysis of Methotrexate and Folic Acid", 1967, The Journal of Biological Chemistry, vol. 242, No. 12, pp. 2933-2938.
Lewis et al., "Receptor-mediated folate uptake is positively regulated by disruption of actin cytoskeleton," Cancer Res. 58(14): 2952-2956 (1998).
Li et al, "Targeted delivery of antisense oligodeoxynucleotides by LPDII," J. Liposome Res. 7(1): 63 (1997).
Li et al. "Local concentration of folate binding protein GP38 in sections of human ovarian carcinoma by concentration of in vitro quantitative autoradiography." J. Nucl. Med. 1996; 37:665-672.
Linder et al., In vitro & in vivo studies with a-and y-isomers of 99′Tc-oxa folate show uptake of both isomers in folate receipt (+) KB Cell Lines J. Nuclear Med. 2000;41(5):470 Suppl.
Liu et al., "Targeted Drug Delivery to Chemoresistant Cells: Folic Acid Derivatization of FdUMP[10] Enhances Cytotoxicity Toward 5-FU-Resistant Human Colorectal Tumor Cells," J. Org. Chem. 66: 5655-5663 (2001).
Lonsdale D, "A Review of the Biochemistry, Metabolism and Clinical Benefits of Thiamin(e) and Its Derivatives," publication, Advance Access Publication, vol. 3, Feb. 2006, pp. 49-59.
Lopes et al (J Chem Soc (Perkin Trans 2) 3:431-440, 1999). *
Lopes, Francisca, et al., "Acyloxymethyl as a Drug Protecting Group. Part 5.1 Kinetics and Mechanism of the Hydrolysis of Tertiary N-Acyloxy-Methylsulfonamides", 1999, J. Chem. Soc., Perkin Trans. 2, pp. 431-439.
Low P.S. et al., "Folate Receptor-Targeted Drugs for Cancer and Inflammatory Diseases," Adv Drug Deliv Rev, 2004;56(8):1055-238.
Lu et al., "Folate receptor-targeted immunotherapy of cancer: mechanism and therapeutic potential," Advanced Drug Delivery Reviews, 2004; vol. 56: 1161-76.
Lu et al., "Folate-targeted enzyme prodrug cancer therapy utilizing penicillin-V amidase and a doxorubicin prodrug," J. Drug Target, 7(1): 43-53 (1999).
Lu, J. Y. And Low, P. S., "Folate targeting of haptens to cancer cell surfaces mediates immunotherapy of syngeneic murine tumors," Cancer Immunol Immunother, 51: 153-162 (2002).
Lu, J. Y. And Low, P. S., "Folate-mediated delivery of macromolecular anticancer therapeutic agents," Adv. Drug Del Rev, 2002; 54(5): 675-693.
Luo et al., "Efficient syntheses of pyrofolic acid and pteroyl azide, reagents for the production of carboxyl-differentiated derivatives of folic acid," J. Am. Chem. Soc., 119: 10004-10013 (1997).
Mack D.O. et al., "The Carboxylation Activity of Vitamin K Analogs with Substitutions at Position 2, 3, or 5," Journal of Biological Chemistry, 1979; vol. 254, pp. 2656-2664.
Mancuso A.J. et al., "Activated Dimethyl Sulfoxide: Useful Reagents for Synthesis," Synthesis, 1981, pp. 165-184.
March Advanced Organic Chemistry, 1992, John Wiley & Sons, 4th Ed. pp. 362-363, 816, 885, and 896.
Mathais et al., "Receptor-mediated targeting of 67Ga-deferoxamine-folate to folate-receptor-positive human KB tumor xenografts," Nucl Med Biol, 26(1): 23-25 (1999).
Mathais et al., "Synthesis of [(99m)Tc]DTPA-folate and its evaluation as a folate-receptor-targeted radiopharmaceutical," Bioconjug Chem, 11(2): 253-257 (2000).
Mathias et al., "Indium-111-DTPA-Folate as a potential folate-receptor-targeted radiopharmaceutical," J. Nucl. Med., 39(9): 1579-1585 (1998).
Mathias et al., "Tumor-Selective Radiopharmaceutical Targeting Via Receptor-Mediated Endocytosis of Gallium-67-Deferoxamine-Folate," J. Nucl. Med, 37(6): 1003-1008 (1996).
Mathias, C. J., "A kit formulation for preparation of [(111)In]In-DTPA-folate, a folate-receptor-targeted radiopharmaceutical," Nucl. Med. Biol., 25(6): 585-587 (1998).
Matsui et al., "Studies on mitomycins. III. The synthesis and properties of mitomycin derivatives," J Antibiot, 21: 189-198 (1968).
McAlinden TP et al., "Synthesis and Biological Evaluation of a Fluorescent Analogue of Folic Acid," Biochemistry, 1991; 30: 5674-81.
McHugh M et al., "Demonstration of a High Affinity Folate Binder in Human Cell Membranes and its Characterization in Cultured human KB Cells," J Biol Chem, 1979; 254(22):11312-8.
Mehvar R "Dextrans for targeted and sustained delivery of therapeutic and imaging agents" [Review] Journal of Controlled Release, 2000;69(1):1-25.
Melani et al., "Targeting of interleukin 2 to human ovarian carcinoma by fusion with a single-chain Fv of antifolate receptor antibody," Cancer Res. 58(18): 4146-4154 (1998).
Melby, E.L. et al, "Entry of Protein Toxins in Polarized Epithelial Cells"; Cancer Research, 1993; 53: 1755-1760.
Mezzanzanica et al. "Human T-lymphocytes targeted against an established human ovarian carcinoma with a bispecific F(ab′)2 antibody prolong host survival in a murine xenograft model" Cancer Res. 1991; 51:5716-5721.
Miotti et al., "Characterization of Human Ovarian Carcinoma-Associated Antigens Defined by Novel Monoclonal Antibodies with Tumor-Restricted Specificity" Int. J. Cancer, 1987;39:297-303.
Mislick et al., "Transfection of folate-polylysine DNA complexes: evidence for lysosomal delivery," Bioconjug. Chem., 6(5): 512-515 (1995).
Mock D.M. et al., "Urinary Biotin Analogs Increase in Humans During Chronic Supplementation: the Analogs are Biotin Metabolites," Am J Physiol Endocrinol Metab, 1997; 272: E83-E85.
Morshed et al., "Folate transport proteins mediate the bidirectional transport of 5-methyltetrahydrofolate in cultured human proximal tubule cells," J. Nutr., 127(6): 1137-1147 (1997).
Nair et al., "Folate analogs altered in the C9-N10 bridge region. 14. 11-Oxahomofolic acid, a potential antitumor agent", Journal of Medical Chemistry, 23: 59-65 (1980).
Nair et al., "Folate analogs altered in the C9-N10 bridge region. 18. Synthesis and antitumor evaluation of 11-oxahomoaminopterin and related compounds," Journal of Medical Chemistry, 24: 1068-1073 (1981).
Nair et al., "Folate analogs altered in the C9-N10 bridge region: 11-thiohomofolic acid," Journal of Medical Chemistry, 22: 850-855 (1979).
Nair et al., "Folate analogs altered in the C9-N10 bridge region: N10-tosylisohomofolic acid and N10-tosylisohomoaminopterin," Journal of Medical Chemistry, 21: 673-677 (1978).
Nair et al., "Folate analogs. 20. Synthesis and antifolate activity of 1′,2′,3′,4′,5′,6′-hexahydrohomotolic acid," Journal of Medical Chemistry, 26: 135-140 (1983).
Nair et al., "Folate analogs. 21. Synthesis and antifolate and antitumor activities of N10-(cyanomethyl)-5,8- dideazafolic acid," Journal of Medical Chemistry, 26: 605-607 (1983).
Nair et al., "Folate analogs. 22. Synthesis and biological evaluation of two analogs of dihydrofolic acid possessing a 7,8-dihydro-8-oxapterin ring system," Journal of Medical Chemistry, 26: 1164-1168 (1983).
Nair et al., "Folate analogues altered in the C9-N10 bridge region. 10-Oxafolic acid and 10-oxaaminopterin," Journal of Medical Chemistry, 19: 825-829 (1976).
Nauta, Jeroen, et al., "Renal and Biliary Abnormalities in a New Murine Model of Autosomal Recessive Polycystic Kidney Disease", 1993, Pediatr. Nephrol. No. 7, pp. 163-172.
Neuss, N. et al., "Vinca Alkaloids. XXX (1). Chemistry of the Deoxyvinblastines (Deoxy-VLB), Leurosine (VLR), and Pleurosine, Dimeric Alkaloids From Vinca," Tetrahedron Letters, No. 7, pp. 783-787 (1968).
Neuzil J et al , "Vitamin E Analogs: A New Class of Multiple Action Agents with Anti-Neoplastic and Anti-Atherogenic Activity," Apoptosis, 2002; vol. 7, pp. 179-187.
Nielsen P. et al., "Phosphates of Riboflavin and Riboflavin Analogs: A Reinvestigation by High-Performance Liquid Chromatography," Analytical Biochemistry, vol. 130, 1983, pp. 359-368.
Nimmo-Smirth R.H. et al., "Some Effects of 2-deaminopteroylglutamic Acid upon Bacterial Growth," J. Gen. Microbial., 1953; 9: 536-544.
Nishikawa, Yuji et al., "Growth Inhibition of Hepatoma Cells Induced by Vitamin K and Its Analogs," Journal of Biological Chemistry, 1995; vol. 270, No. 47, pp. 28304-28310.
Nomura, Makoto et al., "Development of an Efficient Intermediate α-[2-(Trimethylsily1)ethoxy]-2-N-[2- (trimethylsily1)ethoxycarbony1]folic Acid, for the Synthesis of Folate (γ)-Conjugates, and Its Application to the Synthesis of Folate-Nucleoside Congugates," Journal of Organic Chemistry, 2000, vol. 65, pp. 5016-5021.
Nosaka K.et al., "Separate Determination of Anticoccidial Thiamine Analogs by High-performance Liquid Chromatography," ActaA Vitaminol. Et Enzymol, 1984, vol. 6 (2), pp. 137-142.
Oatis et al., "Synthesis of quinazoline analogues of folic acid modified at position 10," Journal of Medical Chemistry, 20: 1393-1396 (1977).
Olsnes S. et al., "Immunotoxins-entry into cells and mechanisms of action," Immunology Today, 1989; vol. 10, No. 9, pp. 291-295.
Pando, O., et al, "First Total Synthesis of Tubulysin B." Org. Lett., 11(24): 5567-5569 (2009).
Pastan et al, eds. "The Pathways of Endocytosis" Endocytosis, Plenum Press, New York 1985;1-40.
Patrick et al., "Folate Receptors as Potential Therapeutic Targets in Choroid Plexus Tumors of Sv40 Transgenic Mice," J. Neurooncol,. 32(2): 111-123 (1997).
Patrick et al., "Intracerebral bispecific ligand-antibody conjugate increases survival of animals bearing endogenously arising brain tumors," Int. J. Cancer, 78(4): 470-79 (1998).
Patterson, Andrew W., et al., "Design, Synthesis, and Biological Properties of Highly Potent Tubulysin D Analogues", 2007, Chem, Eur, J., No. 13, pp. 9534-9541.
Patterson, Andrew W., Hillary M. Peltier, and Jonathan A. Ellman. "Expedient synthesis of N-methyl tubulysin analogues with high cytotoxicity." The Journal of organic chemistry 73(12): 4362-4369 (2008).
Paulos CM et al., "Ligand Binding and Kinetics of Folate Receptor Recycling in Vivo: Impact on Receptor-Mediated Drug Delivery," Molecular Pharmacology, 2004; 66:1406-1414.
Paulos et al., "Folate receptor-mediated targeting of therapeutic and imaging agents to activated macrophages in rheumatoid arthritis," Advanced Drug Delivery Reviews, 2004; vol. 56: 1205-17.
PCT Search Report and Written Opinion for US2008/080948, completed Dec. 4, 2008.
PCT Search Report for PCT/EP2003/011603, completed Feb. 11, 2004.
Peltier, Hillary M., et al., "The Total Synthesis of Tubulysin D," 2006, J. Am. Chem. Soc. vol. 128, pp. 16018-16019.
Peterson et al. "Enzymatic cleavage of peptide-linked radiolabels from immunoconjugates" Bioconjugate Chemistry, 1999;10(4):553-7.
Piontek, Klaus B., et al. "A Functional Floxed Allele of Pkd1 that Can Be Conditionally Inactivated In Vivo", J. Am. Soc. Nephrol. vol. 15, pp. 3035-3043, 2004.
Pizzorno et al. "5,10-Dideazatetrahydrofolic acid (DDATHF) transport in CCRFCEM and MA104 cell lines." J. Biol, Chem., 1993; 268(2):247-258.
Pizzorno G., et al., "Intracellular metabolism of 5,10-didcazatetrahydrofolic acid in human leukemia cell lines," Molecular Pharmacology, 1991, 39 (1), pp. 85-89.
Plante et al., "Polyglutamyl and polylysyl derivatives of the lysine analogues of folic acid and homofolic acid," Journal of Medical Chemistry, 19: 1295-1299 (1976).
Politis I. et al., "The Effect of Various Vitamin E Derivatives on the Urokinase-Plasminogen Activator System of Ovine Macrophages and Neutrophils," British Journal of Nutrition, vol. 89, 2003, pp. 259-265.
Prabhu V. et al., "Arabidopsis dihydropteroate synthase: general properties and inhibition by reaction product and sulfonamides," Phytochem., 1997; 45(1): 23-27.
Prasad et al., "Functional coupling between a bafilomycin A1-sensitive proton pump and a probenecid-sensitive folate transporter in human placental choriocarcinoma cells," Biochim. Biophys. Acta, 1994; 1222(2): 309.
Pratt, A.G. et al. "The Hydrolysis of Mono-, Di, and Triglutamate Derivatives of Folic Acid With Bacterial Enzymes," The Journal of Biological Chemistry, 1968, vol. 243, No. 24, pp. 6367-6372.
Principles of Ion Exchange Chromatography, http://www.separations.us.tosohbioscience.com/ServiceSupport/TechSupport/ResourceCenter/PrinciplesofChromatography/IonExchange, downloaded Dec. 23, 2009.
Punj, V. et al., "Effect of Vitamin D Analog (1α Hydroxy D5) Immunoconjugated to Her-2 Antibody on Breast Cancer," Int. J. Cancer, 2004; 108: 922-929.
Raghavan, Bhooma, et al. "Cytotoxic Simplified Tubulysin Analogues," J. Med. Chem. 51:1530-1533 (2008).
Raghavan, Bhooma, et al., "Cytotoxic Simplified Tubulysin Analogues", 2008, J. Med. Chem., vol. 51, pp. 1530-1533.
Ranastnghe, M. G. et al.; "A Facile Synthesis of Unsymmetrical Thiolsulfonates via Sulfonylation of Mercaptans," Synthetic Communications, 1988; 18(3), pp. 227-232.
Reddy et al., "Optimization of folate-conjugated liposomal vectors for folate receptor-mediated gene therapy," J. Pharm. Sci, 88(11): 1112-1118 (1999).
Reddy et al., "Preclinical evaluation of EC145, a folate-vinca alkaloid conjugate," Cancer Res., 2007; 67:4434-42.
Reddy et al., "Retargeting of viral vectors to the folate receptor endocytic pathway," J Control Release, 74(1-3): 77-82 (2001).
Reddy, J. A., Low, P. S., "Folate-Mediated Targeting of Therapeutic and Imaging Agents to Cancers," Crit. Rev. Ther. Drug Carrier Syst., vol. 15, No. 6, 1998, pp. 587-627.
Regueiro-Ren et al., "Synthesis and Biological Activity of Novel Epothilone Aziridines," Organic Letters, 2001; vol. 3, No. 17: 2693-96.
Remington: The Science & Practice of Pharmacy, 21th Edition (Lippincott Williams & Wilkins, 2005).
Renz P. et al., "Synthesis of 4-Aza-5, 6-diethylbenzimidazole and Biosynthetic Preparation of 4- and 7-Aza-5, 6-dimethylbenzimidazolylcobamide," Z. Nattuforsch, 1997, vol. 52c, pp. 287-291.
Rijnboutt et al., "Endocytosis of GPI-linked membrane folate receptor-alpha," J. Cell Biol., 132(1-2): 35-47 (1996).
Roberts et al., "Folic acid analogs. Modifications in the benzene-ring region. 1. 2′- and 3′-Azafolic acids," Journal of Medical Chemistry, 14: 125-130 (1971).
Roberts et al., "Folic acid analogs. Modifications in the benzene-ring region. 2. Thiazole analogs," Journal of Medical Chemistry, 15: 1310-1312 (1972).
Roberts et al., "Folic acid analogs. Modifications in the benzene-ring region. 3. Neohomofolic and neobishomofolic acids. An improved synthesis of folic acid and its analogs," Journal of Medical Chemistry, 16: 697-699 (1973).
Roberts et al., "Folic acid analogs. Modifications in the benzene-ring region. 4. 3′-Ethyl- and 3′-isopropylfolic acids," Journal of Medical Chemistry, 17: 219-222 (1974).
Rose, William C., "Taxol-Based Combination Chemotherapy and Other in Vivo Preclinical Antitumor Studies", 1993, J. Natl. Cancer Inst. Monographs, No. 15, pp. 47-53.
Ross et al., "Differential regulation of folate receptor isoforms in normal and malignant tissues in vivo and in established cell lines. Physiologic and clinical implications," Cancer, 73(9): 2432-2443, (1994).
Rothberg et al, "Cholesterol controls the clustering of the glycophospholipid-anchored membrane receptor for 5-methyltetrahydrofolate," J. Cell Biol., 111(6): 2931-2938 (1990).
Rothberg et al., "The glycophospholipid-linked folate receptor internalizes folate without entering the clathrin-coated pit endocytic pathway," J. Cell Biol., 110(3): 637-649 (1990).
Rothenberg et al. "Further observations on the folate-binding factor in some leukemic cells" J. Clin. invest. 1971; 50(3):719-726.
Roy et al., "Folate-mediated targeting of T cells to tumors," Advanced Drug Delivery Reviews, 2004; vol. 56: 1219-31.
Roy et al., "Targeting T cells against brain tumors with a bispecific ligand-antibody conjugate," Int. J. Cancer 76(5): 761-66 (1998).
Sabharanjak et al., "Folate receptor endocytosis and trafficking," Advanced Drug Delivery Reviews, 2004; vol. 56: 1099-1109.
Sadasivan et al., "The complete amino acid sequence of a human folate binding protein from KB cells determined from the cDNA," J. Biol. Chem., 1989; 264: 5806-5811.
Sargent D.R. et al., "Antimetabolites of Pantothenic Acid, Ureido-and Carbamoyl-Derivatives," Texas Reports on Biology and Medicine, 1975, vol. 33, No. 3, pp. 433-443.
Sasse F. et al., "Tubulysins, New Cytostatic Peptides from Myxobacteria Acting on Microtubuli Production, Isolation, Physico-Chemical and Biological Properties," The Journal of Antibiotics, 2000; vol. 53, No. 9, pp. 879-885.
Scott J.M, "Preparation and Purification of Pteroic Acid from Pteroylglutamic Acid (Folic Acid)," Methods in Enzymology, 1980, vol. 66, pp. 657-660.
Search Report for Taiwan Patent Application No. 093101735, dated Jul. 14, 2007, 1 page.
Selhub et al. "Renal folate adsorption and the kidney folate binding protein I. Urinary Clearance studies" Am. J Physiol. 252:F750-F756, 1987.
Selhub et al. "Renal folate adsorption and the kidney folate binding protein II. Microinfusion studies" Am. J. Physiol. 252:F757-F760, 1987.
Selhub et al. "The folate binding protein of rat kidney. Purification, properties, and cellular distribution" J. Biol. Chem. 1984;259(10):6601-6606.
Semb J. et al., "Pteroic Acid Derivatives. V. Pteroyl-α-glutamyl-α-glutamylglutamic Acid, Pteroyl-γ-glutamyl-60 -glutamylglutamic Acid, Pteroyl-α-glutamyl-γ-glutamylglutamic Acid," JACS, 1949; 71 (7): 2310-2315.
Senter et al., "Development of a Drug-Release Strategy Based on the Reductive Fragmentation of Benzyl Carbamate Disulfides," J. Org. Chem., 55: 2975-2978 (1990).
Shealy Y.F., "Synthesis and Evaluation of Some New Retinoids for Cancer Chemoprevention," Preventive Medicine, 1989, vol. 18, pp. 624-645.
Shillingford, Jonathan M., et al., "The mTOR Pathway is Regulated by Polycystin-1, and its Inhibition Reverses Renal Cystogenesis in Polycyctic Kidney Disease", Apr. 4, 2006, PNAS. vol. 103, No. 14, pp. 5466-5471.
Shimizu M. et al., "Synthesis and biological activities of new lalpha, 25-dihydroxy-19-norvitamin D3 analogs with modifications in both the A-ring and the side chain," Bioorganic & Medicinal Chemistry, 2006; 14(12): 4277-94.
Shimizu, Kazui, et al., "Novel vitamin D3 antipsoriatic antedrugs: 16-En-22-oxa-1a,25-(OH)2D3 analogs," Bioorganic & Medicinal Chemistry, 2006;14: 1838-1850.
Shoup T.M. et al., "Synthesis of Fluorine-18-Labeled Biotin Derivatives: Biodistribution and Infection Localization," J. Nucl. Med., 1994; 35: 1685-1690.
Sirotnak. "Obligate genetic expression in tumor cells of a fetal membrane property mediating "Folate" transport: biological significance and implications for improved therapy of human cancer" Cancer Res., 1985;45(9):3992-4000.
Skinner W.A. et al., "Structure-Activity Relations in the Vitamin E Series. II. Derivatives of α-Tocopherol Substituted at the 5-Methyl Group," J. Med. Chem., 1969; 12 (1): 64-66.
Smart et al., "Clustered folate receptors deliver 5-methyltetrahydrofolate to cytoplasm of MA104 cells," J. Cell Biol., 134(5): 1169-1177 (1996).
Smart et al., "Protein kinase C activators inhibit receptor-mediated potocytosis by preventing internalization of caveolae," J. Cell Biol., 124(3): 307-313 (1994).
Speckamp et al (Tetrahedron 56:3817-3856, 2000). *
Spry C. et al., "A Class of Pantothenic Acid Analogs Inhibits Plasmodium Falciparum Pantothenate Kinase and Represses the Proliferation of Malaria Parasites," Antimicrobial Agents and Chemotherapy, 2005; 49(11): 4649-4657.
Stein et al. "Normal tissue reactivity of four anti-tumor monoclonal antibodies of clinical interest" Int. J. Cancer 1991;47(2):163-169.
Steinberg, G. et al., "Synthesis and Evaluation of Pteroic Acid-Conjugated Nitroheterocyclic Phosphoramidates as Folate Receptor-Targeted Alkylating Agents," J. Med. Chem. 44: 69-73 (2001).
Steinmetz et al (Angew Chem Int Ed 43:4888-4892, 2004). *
Steinmetz, H. et al., "Isolation, Crystal and Solution Structure Determination, and Biosynthesis of Tubulysins-Powerful Inhibitors of Tubulin Polymerization from Microbacteria", Angew. Chem. Int. Ed., 2004, No. 43, pp. 4888-4892.
Takahata Y. et al., "Synthesis, Properties and Microbiological Activity of Hydrophobic Derivatives of Vitamin B12," J. Nutr. Sci. Vitaminol., 1995, vol. 41, pp. 515-526.
Takasu, H. et al., "c-Fos protein as a target of anti-osteoclastogenic action of vitamin D, and synthesis of new analogs," The Journal of Clinical Investigation, 2006; vol. 116, No. 2, pp. 528-535.
Takeda, K. et al., "A Synthesis of a New Type of Alkoxycarbonylating Reagents from 1,1-Bis[6-(trifluoromethyl)benzotriazolyl] Carbonate (BTBC) and Their Reactions," Sythesis, 1987; 6: 557-560.
Tanaka et al. "Preparation and characterization of a disulfide-linked bioconjugate of annexin V with the B-chain of urokinase: an improved fibrinolytic agent targeted to phospholipid-containing thrombi" Biochemistry, 1996;35(3):922-9.
Temple et al., "Synthesis of pseudo cofactor analogs as potential inhibitors of the folate enzymes," Journal of Medical Chemistry, 25: 161-166 (1982).
Thaden et al. "Photoaffinity behavior of a conjugate of oligonucleoside methylphosphonate, rhodamine, and psoralen in the presence of complementary oligonucleotides" Bioconjugate Chemistry, 1993;4(5):386-94.
Theti, D. S. et al., "Selective delivery of CB300638, a cyclopenta[g]quinazoline-based thymidylate synthase inhibitor into human tumor cell lines overexpres sing the alpha-isoform of the folate receptor," Cancer Res, 2003; 63(13): 3612-3618.
Toffoli et al. "Expression of folate binding protein as a prognostic factor for response to platinum-containing chemotherapy and survival in human ovarian cancer" int. J. Cancer (Pred. Oncol) 1998; 79:121-126.
Toffoli et al., "Overexpression of folate binding protein in ovarian cancers" Int. J. Cancer 74(2): 193-198 (1997).
Toraya T. et al., "Immobilized Derivatives of Vitamin B12 Coenzyme and Its Analogs," Methods in Enzymology, vol. 67, pp. 57-66, 1980.
Toraya T. et al., "The Synthesis of Several Immobilized Derivatives of Vitamin B12 Coenzyme and Their Use as Affinity Adsorbents for a Study of Interactions of Diol Dehydrase with the Coenzyme," The Journal of Biological Chemistry, 1990; vol. 255, No. 8, pp. 3520-3525.
Traciiewsky D., "Antihypertensive Effect of Riboflavin Analogs in Rats with Mineralocorticoid-Induced Hypertension," Hypertension, 1981; vol. 3, No. 1, pp. 75-80.
Trunch A. et al., "Temperature-sensitive differential affinity of TRAIL for its receptors. DR5 is the highest affinity receptor," J Biol Chem, 2000; 275(30):23319-25.
Turek et al., "Endocytosis of folate-protein conjugates: ultrastructural localization in KB cells," J. Cell Sci. 106: 423-430 (1993).
Turk et al., "Characterization of a novel pH-sensitive peptide that enhances drug release from folate-targeted liposomes at endosomal pHs," Biochim Biophys Acta, 1559(1): 56-68 (2002).
U.S. Appl. No. 12/666,712, filed Dec. 24, 2009, Leamon et al.
U.S. Appl. No. 12/775,824, filed May 7, 2010, Green et al.
U.S. Appl. No. 60/590,580, filed Jul. 23, 2004, Vlahov et al.
U.S. Appl. No. 60/946,092, filed Jun. 25, 2007, Vlahov et al.
U.S. Appl. No. 60/982,595, filed Oct. 25, 2007, Vlahov et al.
U.S. Appl. No. 61/036,176, filed Mar. 13, 2008, Vlahov et al.
U.S. Appl. No. 61/036,186, filed Mar. 13, 2008, Vlahov et al.
Ueda M. et al., "Effect of Vitamin B12 Derivatives on Urinary Excretion of Methylmalonic Acid in Liver Diseases," Acta Med. Okayama, 1970; vol. 24, pp. 365-372.
Varma, R. et al., "GPI-anchored proteins are organized in submicron domains at the surface," Nature, 394(6695): 798-801 (1998).
Verwey, J., "Mitomycin C-Induced Renal Toxicity, a Dose-Dependent Side Effect?," Eur J Cancer Clin Onco, 1987; vol. 23, No. 2, pp. 195-199.
Vesely D.L. et al., "Biotin Analogs Activate Guanylate Cyclase," Molecular and Cellular Biochemistry, 1984; vol. 60, pp. 109-114.
Vlahov I.R. et al., "Design and regioselective synthesis of a new generation of targeted chemotherapeutics. Part 1: EC145, a folic acid conjugate of desacetylvinblastine monohydrazide," Bioorg Med Chem Lett, 2006; 16(19):5093-6.
Vogel et al., "Peptide-Mediated Release of Folate-Targeted Liposome Contents From Endosomal Compartments," J. Am. Chem. Soc., 1996; 118(7): 1581-1586.
Vyas D. et al., "A practical synthesis of mitomycin a and its analogs," J Org Chem, 1986; 31:4307-4309.
Wang et al., "Delivery of antisense oligodeoxyribonucleotides against the human epidermal growth factor receptor into cultured KB cells with liposomes conjugated to folate via polyethylene glycol," Proc. Natl. Acad. Sci. USA, 92(8): 3318-3322 (1995).
Wang et al., "Design and synthesis of [111In]DTPA-folate for use as a tumor-targeted radiopharmaceutical," Bioconjug Chem., 8(5): 673-679 (1997).
Wang et al., "Synthesis, purification, and tumor cell uptake of 67Ga-deferoxamine-folate, a potential radiopharmaceutical for tumor imaging," Bioconj. Chem., 1996; 7(1): 56-62.
Wang S. et al., "Folate-mediated targeting of antineoplastic drugs, imaging agents, and nucleic acids to cancer cells," J. Control Rel, 1998; 53(1-3): 39-48.
Wang, Xiu-Fang et al., "Vitamin E Analogs Trigger Apoptosis in HER2/erbB2-Overexpressing Breast Cancer Cells by Signaling Via the Mitochondrial Pathway," Biochemical and Biophysical Research Communication, 2005; vol. 326, pp. 282-289.
Wang, Zhiyong, et al., "Structure-Activity and High-Content Imaging Analysis of Novel Tubulysins", 2007, Chem Biol. Drug Des., No. 70, pp. 75-86.
Weinstock et al., "Folic acid analogs. II. p-{[(2,6-Diamino-8-purinyl)methyl]amino}-benzoyl-L-glutamic acid and related compounds," Journal of Medical Chemistry, 13: 995-997 (1970).
Weitman et al., "Cellular localization of the folate receptor: potential role in drug toxicity and folate homeostasis," Cancer Res., 1992; 52(23): 6708-6711.
Weitman et al., "Distribution of the folate receptor GP38 in normal and malignant cell lines and tissues," Cancer Res., 1992; 52(12): 3396-3401.
Westerhof et al. "Membrane transport of natural folates and antifolate compounds in murine L1210 Leukemia cells: role of carrier-and receptor mediated transport systems" Cancer Res. 1991;51:5507-5513.
Westerhof G.R. et al., "Carrier- and Receptor-Mediated Transport of Folate Antagonists Targeting Folate-Dependent Enzymes: Correlates of Molecular-Structure and Biological Activity," Molecular Pharmacology, 1995, 48, pp. 459-471.
Westerhof GR et al., "A photoaffinity analogue of folic acid as a probe for the identification and function of a membrane folate binding protein (mFBP) in human CCRF-CEM leukemia cells," Proceedings of the American Association for Cancer Research, 1991; 32:328.
Weygand, et al., Chemishe. Berichte (1950) 83, 460-467.
Wiener et al., "Targeting dendrimer-chelates to tumors and tumor cells expressing the high-affinity folate receptor," Invest. Radiol. 32(12): 748-54 (1997).
Wikipedia, Analog (Chemistry), http://en.wikipedia.org/wiki/Analog-(chemistry), downloaded Dec. 16, 2009.
Wikipedia, Complex (Chemistry), http://en.wikipedia.org/wiki/Complex-(chemistry), downloaded Dec. 23, 2009.
Wikipedia, Conjugate, http://en.wikipedia.org/wiki/Conjugate, downloaded Dec. 17, 2009.
Wikipedia, Derivative (Chemistry), http://en.wikipedia.org/wiki/Derivative-(chemistry), downloaded Dec. 16, 2009.
Wikipedia, Folic acid, http://en.wikipedia.org/wiki/Folic-acid, downloaded Apr. 7, 2009.
Wikipedia, Functional analog, http://en.wikipedia.org/wiki/Functional-analog, downloaded Apr. 7, 2009.
Wikipedia, List of Purification Methods in Chemistry, http://en.wikipedia.org/wiki/List-of-purification-methods-in-chemistry, downloaded Dec. 16, 2009.
Wikipedia, Solution, http://en.wikipedia.org/wiki/Solution, downloaded Dec. 17, 2009.
Wikipedia, Structural analog, http://en.wikipedia.org/wiki/Structural-analog, downloaded Apr. 7, 2009.
Williams et al. "Renal tubular transport of folic acid and methotrexate in the monkey" Am. J. Physiol 1982; 242(5):F484-490.
Wu M. et al., "Clustering of GPI-anchored folate receptor independent of both cross-linking and association with caveolin," J. Membr. Biol. 159(2): 137-147 (1997).
Wu, Shih Hsiung, Zhi Wei Guo, and Charles J. Sih. "Enhancing the enantioselectivity of Candida lipase-catalyzed ester hydrolysis via noncovalent enzyme modification." Journal of the American Chemical Society 112(5): 1990-1995 (1990).
Zhao et al., "Tumor-selective targeted delivery of genes and antisense oligodeoxyribonucleotides via the folate receptor," Advanced Drug Delivery Reviews, 2004; vol. 56: 1193-1204.
Zimmer H. et al., "Potential anticancer agents V., Synthesis of folic acid and pteroic acid analogs derived of caffeine and theophylline," Arzneimittelforschung, 1966, 16(4), pp. 541-545.
Zimmerman, J., "Folic acid transport in organ-cultured mucosa of human intestine. Evidence for distinct carriers," Gastroenterol. 99(4): 964-972 (1990).

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9745341B2 (en) 2007-10-25 2017-08-29 Endocyte, Inc. Tubulysins and processes for preparing
US20160016993A1 (en) * 2013-03-01 2016-01-21 Endocyte, Inc. Process for preparing tubulysins
US9771391B2 (en) * 2013-03-01 2017-09-26 Endocyte, Inc. Process for preparing tubulysins

Also Published As

Publication number Publication date
EP2209374A1 (en) 2010-07-28
AU2008316835A1 (en) 2009-04-30
EP2209374A4 (en) 2011-08-17
WO2009055562A1 (en) 2009-04-30
US9745341B2 (en) 2017-08-29
CA2703491A1 (en) 2009-04-30
CA2703491C (en) 2017-06-13
IL205253A0 (en) 2010-12-30
CN101909441A (en) 2010-12-08
JP2011500835A (en) 2011-01-06
IL205253A (en) 2015-07-30
US20160108085A1 (en) 2016-04-21
CN101909441B (en) 2015-05-13
US20100240701A1 (en) 2010-09-23
JP2017061469A (en) 2017-03-30
JP2014240387A (en) 2014-12-25
JP6077493B2 (en) 2017-02-08
EP2209374B1 (en) 2014-12-03
AU2008316835B2 (en) 2015-07-16

Similar Documents

Publication Publication Date Title
US9745341B2 (en) Tubulysins and processes for preparing
JP7079355B2 (en) PSMA-binding ligand-linker conjugate and usage
CN109316605B (en) Folate receptor binding ligand-drug conjugates
JP2019081778A (en) Drug delivery conjugates, and methods for treating diseases caused by psma expressing cells
JP5907513B2 (en) Tubulysine analogs
US20140107316A1 (en) Drug delivery conjugates containing unnatural amino acids and methods for using
US20090246211A1 (en) Molecular constructs suitable for targeted conjugates
KR20080070060A (en) Alpha;-HELIX MIMETICS AND METHOD RELATING TO THE TREATMENT OF CANCER STEM CELLS
CN106279352B (en) Derivative of dolastatin 10 and application thereof
AU2022204711A1 (en) Peptidic linkers and cryptophycin conjugates, useful in therapy, and their preparation
JP2005501121A (en) Hydrazinopeptoids and their use for treating cancer
US20160303251A1 (en) Conjugates of garftase inhibitors
KR20080091103A (en) Compound for the inhibition of apoptosis
CA2945367A1 (en) Drug delivery conjugates for treating resistant cancer and for use in combination therapy
AU2018329951B2 (en) Process for the preparation of tubulysins and intermediates thereof
AU2013203148A1 (en) Tubulysins and processes for preparing
WO2015069766A1 (en) Dupa-indenoisoquinoline conjugates
US11518786B2 (en) Third generation tubulysin analogues and process of preparation thereof
WO2022175222A1 (en) Cryptophycin compounds and conjugates thereof
Bielawski et al. Synthesis and Cytotoxic Activity of Novel Amidine Analogues of Bis (2‐chloroethyl) amine
WO2023144379A1 (en) High-affinity ligands of fibroblast activation protein for targeted delivery applications
WO2021101407A1 (en) Conjugate monomethyl auristatin e to obtain a composition for treatment of prostate cancer
JP2021536475A (en) Alternative process for the preparation of tubelysine and its intermediates
MX2008006684A (en) Compounds for the inhibition of apoptosis

Legal Events

Date Code Title Description
AS Assignment

Owner name: ENDOCYTE, INC., INDIANA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:VLAHOV, IONTCHO RADOSLAVOV;WANG, YU;LEAMON, CHRISTOPHER PAUL;REEL/FRAME:021917/0823

Effective date: 20081124

STCF Information on status: patent grant

Free format text: PATENTED CASE

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YR, SMALL ENTITY (ORIGINAL EVENT CODE: M2551); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

Year of fee payment: 4

FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20231117